WO2021185851A1 - Vaccine compositions for hiv prevention and treatment - Google Patents

Vaccine compositions for hiv prevention and treatment Download PDF

Info

Publication number
WO2021185851A1
WO2021185851A1 PCT/EP2021/056710 EP2021056710W WO2021185851A1 WO 2021185851 A1 WO2021185851 A1 WO 2021185851A1 EP 2021056710 W EP2021056710 W EP 2021056710W WO 2021185851 A1 WO2021185851 A1 WO 2021185851A1
Authority
WO
WIPO (PCT)
Prior art keywords
hiv
peptide
faba
clade
mucosal
Prior art date
Application number
PCT/EP2021/056710
Other languages
French (fr)
Inventor
Morgane Bomsel
Daniela Tudor
Pierre TUFFERY
Julien Rey
Original Assignee
INSERM (Institut National de la Santé et de la Recherche Médicale)
Centre National De La Recherche Scientifique (Cnrs)
Université de Paris
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by INSERM (Institut National de la Santé et de la Recherche Médicale), Centre National De La Recherche Scientifique (Cnrs), Université de Paris filed Critical INSERM (Institut National de la Santé et de la Recherche Médicale)
Publication of WO2021185851A1 publication Critical patent/WO2021185851A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/005Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from viruses
    • C07K14/08RNA viruses
    • C07K14/15Retroviridae, e.g. bovine leukaemia virus, feline leukaemia virus human T-cell leukaemia-lymphoma virus
    • C07K14/155Lentiviridae, e.g. visna-maedi virus, equine infectious virus, FIV, SIV
    • C07K14/16HIV-1 ; HIV-2
    • C07K14/162HIV-1 ; HIV-2 env, e.g. gp160, gp110/120, gp41, V3, peptid T, CD4-Binding site
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/54Medicinal preparations containing antigens or antibodies characterised by the route of administration
    • A61K2039/541Mucosal route

Definitions

  • the invention is in the infection field. More particularly, the invention relates to novel (poly) peptides relating to HIV are disclosed, as are vaccine compositions containing such (poly) peptides.
  • the compounds and compositions of the invention are of use in prevention and treatment of HIV, and in particular for inducing mucosal immunity to HIV.
  • HIV-1 infection is mainly initiated at mucosal sites during sexual intercourses, and IgA can efficiently prevent HIV-1 infection and mucosal reservoir establishment.
  • IgA can efficiently prevent HIV-1 infection and mucosal reservoir establishment.
  • mucosal IgAs specific for HIV-1 envelope gp41-subunit conserved epitopes, naturally raise in 1% of individuals upon sexual exposure to HIV-1 who remain persistently uninfected. These individuals, referred to as Exposed SeroNegatives (ESN), remain IgG seronegative but raise gp41 -specific mucosal IgA that constitute a main correlate of protection (reviewed in [1] [2] [5]).
  • ESN IgA are able to inhibit HIV-1 mucosal penetration by transcytosis across epithelial barriers and to neutralize CD4 + T-cell infection against Clade B HIV-1 [2] [6] [7]
  • These IgA detected mainly in vaginal washes and urine, recognize the conserved regions of the transmembrane subunit gp41 of HIV-1 envelope [6] [2, 7]
  • ESN IgAs are affinity matured with extensive somatic hypermutations, and have extended heavy-chain third complementary determining regions, (CDRH3) [2], characteristics similar to those found for IgGs in natural HIV-1 infection and that increase breadth and potency in vitro [8] [9] [10]
  • CDRH3 third complementary determining regions
  • ESN protective gp41 -specific IgAs at mucosal level evidences an efficient “natural” vaccination. Furthermore, this IgA-driven immune response in ESN questions about the protective mechanisms of a humoral response involving only mucosal IgA, and, at the molecular level, about the participation of the isotype specific constant regions of the antibody in ESN IgA efficacy.
  • Antibodies have been historically described as being composed of two distinct structurally independent domains, namely the variable regions, forming the paratope, responsible for antigen binding, and the heavy chain constant regions mediating effector functions cells such as antibody dependent cell cytotoxicity (ADCC) and antibody dependent phagocytosis (ADCP).
  • ADCC antibody dependent cell cytotoxicity
  • ADCP antibody dependent phagocytosis
  • CH Ab heavy chain constant region
  • CH3 and CH2 participate in Fc- receptor binding on effector cells to mediate ADCC or ADCP whereas the CHI domain, separated from the two others by the hinge region, is part of the Fab region of the Ab and closer to the paratope formed by the variable heavy (VH) and light (VL) chains.
  • VH variable heavy
  • VL variable light
  • Which CH domain(s) participates in modulation of Ab specificity and affinity remains unclear and whether the CHI itself, due to its proximity to the VH and VL regions, plays a determinant role in Ab affinity and functions has not been studied.
  • the role of constant regions has only been evaluated by comparing blood derived antibodies expressing identical V regions in different heavy chain constant context and thus addressing a blood raised B-cell epitope.
  • mucosal immunity is highly compartmentalized [17] resulting in the induction of mucosal antibody recognizing B cell epitope that differ from systemic one. Whether the antibody isoptype also affects Ab specificity and affinity and even function of mucosal Abs in a similar manner than blood derived Abs remains unknown.
  • HIV-I envelope glycoproteins Most of the vaccine approaches against HIV-I have targeted the viral envelope (Env) glycoproteins because they are the major surface antigens expressed on virions and by HIV-I infected cells.
  • the native HIV-I envelope glycoprotein is a heterotrimer containing three gpl20 proteins non-covalently associated with three gp41 glycoproteins.
  • the native envelope glycoprotein complex such as the gpl20/gp41.
  • many potential cross-reactivities events may occur leading to possible harmful autoimmune phenomena.
  • the inventor took advantage of two ESN mucosal anti-HIV-1 Fab-IgAl (FabA) previously characterized [2], that were isotype switched into Fab-IgGl (FabG).
  • FabA and its respective isotype switched FabG were compared for HIV-1 envelope binding, epitope recognition, and anti- viral activities against the main HIV-1 strains present worldwide, namely HIV-1 of Clades B, A and C, and their conformational epitopes on gp41 were determined using in silico analyses.
  • These comparative analyses result in a molecular understanding of the recognition of different epitopes on HIV-1 envelope, depending on the isotype in correlation with different affinities and antiviral functions.
  • the inventors used a reverse vaccinology approach based on the Fab conformational epitopes to generate a (poly) peptide that is useful in therapeutic approaches against HIV infection.
  • the invention provides a peptide comprising or consisting of the consecutive sequence of amino acids (SEQ IDN°1): LWNWFDISAASI
  • the peptide is useful in preventive and curative treatments of HIV infections.
  • the invention provides a therapeutic composition comprising a peptide of the invention, wherein the peptide is used for its direct neutralizing properties against an HIV infection, namely by clade A, B and C virus.
  • the peptide is used preferably systemically, as an antiviral agent.
  • the invention provides a therapeutic composition comprising a peptide of the invention, wherein the peptide is used for its antigenic properties for inducing a protective immunity against an HIV infection.
  • nucleic acids encoding the peptides of the invention, vectors comprising said nucleic acids and host cells comprising said vectors and nucleic acids.
  • compositions comprising a peptide, or nucleic acid of the invention.
  • said composition is a vaccine composition.
  • said composition is suitable for mucosal administration.
  • said composition may be suitable for administration to the nasal, rectal or vaginal mucosa.
  • said vaccine composition additionally comprises one or more adjuvants.
  • compositions of the invention for use in the preventive or therapeutic treatment of HIV or AIDS.
  • the composition may be for administration to a mucosal surface, or any other administrative route such as those described below.
  • the composition may be for administration in a prime-boost regimen such as those described below, such as a regimen comprising administration of full-length HIV-1 gp41.
  • a peptide, nucleic acid or composition of the invention for use in a method of inducing a mucosal immune response to HIV-1 n some embodiments, said method comprises administering said peptide, nucleic acid or composition to a subject, in particular a subject in need thereof.
  • the peptide, nucleic acid or composition may be administered to a mucosal surface of a subject, for example the nasal, rectal or vaginal mucosa.
  • Various administration regimens are described herein and encompassed within the scope of the invention.
  • a peptide, nucleic acid or composition of the invention for use in a method of inducing a mixed mucosal IgGl and IgA2 response to HIV-1, the method comprising administration of at least a peptide comprising the peptidic sequence of the invention or nucleic acids encoding said peptides, or a composition comprising said peptides or nucleic acids.
  • a peptide, nucleic acid or composition of the invention for use in the preparation of a medicament for the preventive treatment of HIV infection, in particular HIV- 1 infection, or AIDS.
  • compositions of the invention comprising administration of a composition of the invention to an individual in need thereof.
  • said composition is administered to a mucosal surface of a subject, for example the nasal, rectal or vaginal mucosa.
  • administration regimens are described herein and encompassed within the scope of the invention.
  • a composition of the invention comprising administration of a composition of the invention to an individual in need thereof.
  • said composition is administered to a mucosal surface of a subject, for example the nasal, rectal or vaginal mucosa.
  • Various administration regimens are described herein.
  • Polypeptides include amino acid sequences modified either by natural processes, such as post-translational processing, or by chemical modification techniques which are well known in the art. Such modifications are well described in basic texts and in more detailed monographs, as well as in a voluminous research literature.
  • Modifications can occur anywhere in a polypeptide, including the peptide backbone, the amino acid side- chains and the amino or carboxyl termini, it will be appreciated that the same type of modification may be present in the same or varying degrees at several sites in a given polypeptide. Also, a given polypeptide may contain many types of modifications. Polypeptides may be branched as a result of ubiquitination, and they may be cyclic, with or without branching. Cyclic, branched and branched cyclic polypeptides may result from natural posttxans!ational processes or may be made by synthetic methods.
  • Modifications include acetylation, acylation, ADP-ribosylation, araidation, covalent attachment of flavin, covalent attachment of a heme moiety, covalent attachment of a nucleotide or nucleotide derivative, covalent attachment of a lipid or lipid derivative, covalent attachment of phosphatidyl inositol, cross-linking, cyclization, disulfide bond formation, demethylation, formation of covalent cross-links, formation of cystine, formation of pyroglutamate, formylation, gamma-carboxy!ation, glycosylation, GPI anchor formation, hydroxy!
  • Variants of the peptide disclosed herein comprise peptides sequences of SEQ ⁇ DN°1 wherein 1 to 2 amino acids are substituted with other amino acids resulting in conservative substitutions. Exemplary conservative substitutions are listed below.
  • the peptide vaccines of the invention may be formulated into a pharmaceutical form, preferably in combination with a pharmaceutically acceptable carrier.
  • suitable for injectable use include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions in all cases the form must be sterile and must be fluid to the extent that easy syringability exists. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms, such as bacteria and fungi.
  • the carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (e.g., glycerol, propylene glycol, and liquid polyethylene glycol, and the like), suitable mixtures thereof, and/or vegetable oils.
  • polyol e.g., glycerol, propylene glycol, and liquid polyethylene glycol, and the like
  • suitable mixtures thereof e.g., vegetable oils
  • vegetable oils e.g., glycerol, propylene glycol, and liquid polyethylene glycol, and the like
  • suitable mixtures thereof e.g., glycerol, propylene glycol, and liquid polyethylene glycol, and the like
  • vegetable oils e.g., glycerol, propylene glycol, and liquid polyethylene glycol, and the like
  • Proper fluidity may be maintained, for example, by the use of a coating, such as lecithin, by the maintenance of the required particle size in the case of dispersion
  • isotonic agents for example, sugars or sodium chloride.
  • Prolonged absorption of the injectable compositions can be brought about by the use in the compositions of agents delaying absorption, for example, aluminum monostearate and gelatin.
  • aqueous solutions For parenteral administration in an aqueous solution, for example, the solution should be suitably buffered if necessary and the liquid diluent first rendered isotonic with sufficient saline or glucose.
  • aqueous solutions are especially suitable for intravenous, intraarterial, intramuscular, subcutaneous, intratumoral and intrap eritoneal administration.
  • sterile aqueous media that can be employed will be known to those of skill in the art in light of the present disclosure.
  • one dosage may be dissolved in I ml of isotonic NaCl solution and either added to 1000 ml of hypodermoclysis fluid or injected at the proposed site of infusion, (see for example, (see for example, Remington: The Science and Practice of Pharmacy, 21 st Edition, Lippincot and Williams, 2005). Some variation in dosage will necessarily occur depending on the condition of the subject being treated. The person responsible for administration will, in any event, determine the appropriate dose for the individual subject. Moreover, for human administration, preparations should meet sterility, pyrogenicity, general safety and purity standards as required by FDA Office of Biologies standards.
  • Sterile injectable solutions are prepared by incorporating the active compounds in the required amount in the appropriate solvent with various of the other ingredients enumerated above, as required, followed by filtered sterilization.
  • dispersions are prepared by incorporating the various sterilized active ingredients into a sterile vehicle which contains the basic dispersion medium and the required other ingredients from those enumerated above.
  • the preferred methods of preparation are vacuum-drying and freeze-drying techniques which yield a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.
  • compositions disclosed herein may be formulated in a neutral or salt form.
  • Pharmaceutically-acceptable salts include the acid addition salts and which are formed with inorganic acids such as, for example, hydrochloric or phosphoric acids, or such organic acids as acetic, oxalic, tartaric, mandelic, and the like. Salts formed with the free carboxyl groups can also be derived from inorganic bases such as, for example, sodium, potassium, ammonium, calcium, or ferric hydroxides, and such organic bases as isopropylamine, trimethylamine, histidine, procaine and the like.
  • solutions Upon formulation, solutions will be administered in a manner compatible with the dosage formulation and in such amount as is therapeutically effective.
  • the formulations are easily administered in a variety of dosage forms such as injectable solutions, drug release capsules and the like.
  • carrier includes, without limitation, solvents, dispersion media, vehicles, coatings, diluents, antibacterial and antifungal agents, isotonic and absorption delaying agents, buffers, carrier solutions, suspensions, colloids, liposomes and virosomes such as those described in Bomsel et al (2011) Immunity 34: 269-280.
  • solvents dispersion media, vehicles, coatings, diluents, antibacterial and antifungal agents, isotonic and absorption delaying agents, buffers, carrier solutions, suspensions, colloids, liposomes and virosomes
  • Supplementary active ingredients can also be incorporated into the compositions.
  • phrases ‘pharmaceutically-acceptable’ or ‘pharmacologically-acceptable’ refers to molecular entities and compositions that do not produce an allergic or similar untoward reaction when administered to a human.
  • the preparation of an aqueous composition that contains a protein as an active ingredient is well understood in the art.
  • such compositions are prepared as injectables, either as liquid solutions or suspensions; solid forms suitable for solution in, or suspension in, liquid prior to injection can also be prepared.
  • the vaccine composition will preferably contain an agent that enhances the protective efficacy of the vaccine, such as an adjuvant.
  • adjuvants include any compound or compounds that act to increase a protective immune response to the peptide antigen, thereby reducing the quantity of antigen necessary in the vaccine, and/or the frequency of administration necessary to generate a protective immune response.
  • Adjuvants can include for example, emulsifiers; muramyl dipeptides; avridine; aqueous adjuvants such as aluminum hydroxide; oxygen-containing metal salts; chitosan-based adjuvants, and any of the various saponins, oils, and other substances known in the art, such as Ampfaigen, LPS, bacterial cell wall extracts, bacterial DNA, CpG sequences, synthetic oligonucleotides and combinations thereof (Schijns et al (2000) Curr. Opin. Immunol, 12:456), Mycohacterialplilei ( phlei) cell wall extract ( CWE) (U.S. Patent No. 4,744,984), M.
  • M-D A M-DNA-M phlei ceil wall complex
  • LT heat-labile enterotoxin
  • CT cholera toxin
  • CB cholera toxin B subunit
  • Compounds which can serve as emulsifiers include natural and synthetic emulsifying agents, as well as anionic, cationic and nonionic compounds.
  • Oxygen-containing metal salts include salts of Al, K, Ca, Mg, Zn, Ba, Na, Li, B, Be, Fe, Si, Co, Cu, Ni, Ag, Au, and Cr which are sulphates, hydroxides, phosphates, nitrates, iodates, bromates, carbonates, hydrates, acetates, citrates, oxalates, and tartrates, and mixed forms thereof, including aluminium hydroxide, aluminium phosphate, aluminium sulphate, potassium aluminium sulphate, calcium phosphate, Maalox (mixture of aluminium hydroxide and magnesium hydroxide), beryllium hydroxide, zinc hydroxide, zinc carbonate, zinc chloride, and barium sulphate
  • anionic emulsifying agents include, for example, the potassium, sodium and ammonium sails of lauric and oleic acid, the calcium., magnesium and aluminum salts of fatty acids, and organic sulfonates such as
  • Synthetic cationic agents include, for example, cetyltrhethylammonlum bromide, while synthetic nonionic agents are exemplified by glycerylesters (e.g., glyceryl monostearate), polyoxyethylene glycol esters and ethers, and the sorbitan fatty acid esters (e.g., sorbitan monopalmitate) and their polyoxyethylene derivatives (e.g., polyoxyethylene sorbitan. monopalmitate).
  • Natural emulsifying agents include acacia, gelatin, lecithin and cholesterol.
  • Suitable adjuvants can be formed with an oil component, such as a single oil, a mixture of oils, a water-in-oil emulsion, or an oil-in- water emulsion.
  • the oil can be a mineral oil, a vegetable oil, or an animal oil.
  • .Mineral oils are liquid hydrocarbons obtained from petrolatum via a distillation technique, and are also referred to in the art as liquid paraffin, liquid petrolatum,, or white mineral oil.
  • Suitable animal oils include, for example, cod liver oil, halibut oil, menhaden oil, orange roughy oil and shark liver oil, ail of which are available commercially.
  • Suitable vegetable oils include, for example, canola oii, almond oil, cottonseed oil, com oil, olive oil, peanut oil, safflower oii, sesame oil, soybean oil, and the like.
  • FCA Freund's Complete Adjuvant
  • FI A Freund's incomplete Adjuvant
  • FCA and FIA are water-in-mineral oil emulsions; however, FCA also contains a killed Mycobacterium sp.
  • Particularlay preferred adjuvants for mucosal vaccines include galactosyl ceramide (GalCer), as described in Lee et al (2011) Vaccine 29: 417-425.
  • Immunomodulatory cytokines can also be used in the vaccine compositions to enhance vaccine efficacy, for examplean adjuvant,
  • Non-limiting examples of such cytokines include interferon alpha (IFN-a), interleukin-2 (IL-2), and granulocyte rnacrophage-colony stimulating factor (GM— CSF), or combinations thereof.
  • GM-CSF is preferred.
  • the immunogenic compositions of the invention are ideally administered to a subject in advance of HIV infection, or evidence of HIV infection, or in advance of any symptom due to AIDS, especially in high-risk subjects.
  • the prophylactic administration of the immunogenic compositions can serve to provide protective immunity of a subject against HIV-1 infection or to prevent or attenuate the progression of AIDS in a subject already infected with HIV-1.
  • the immunogenic compositions can serve to ameliorate and treat AIDS symptoms and are advantageously used as soon after infection as possible, preferably before appearance of any symptoms of AIDS but may also be used at (or after) the onset of the disease symptoms.
  • Administration may be via a parenteral or non-parenteral route.
  • Routes of administration will vary, naturally, with the location and nature of the disease, and include, e.g. intravenous, intrarterial, intradermal, transdermal, intramuscular, mucosal subcutaneous, percutaneous, intratracheal, intraperitoneal, perfusion and lavage.
  • administration is via a mucosal route, for example via a nasal, oral (via the mucosa of the digestive system), vaginal, buccal, rectal, sublingual, ocular, urinal, pulmonal or otolar (vie the ear) route.
  • an exemplary formulation may be a nasal spray, lavage, drop or squirt system such as the Bidose Liquid from Aptar, Pfeffer Group or the Accuspray from Becton Dickinson (see Brandztaeg, 2011, AJRCCM, Bitter et al (2011) CurrProbl Dermatol 40: 20-35).
  • squirt system such as the Bidose Liquid from Aptar, Pfeffer Group or the Accuspray from Becton Dickinson (see Brandztaeg, 2011, AJRCCM, Bitter et al (2011) CurrProbl Dermatol 40: 20-35).
  • a unit dose is defined as containing a predetermined-quantity of the therapeutic composition comprising a lentiviral vector of the present invention.
  • the quantity to be administered, and the particular route and formulation, are within the skill of those in the clinical arts.
  • a unit dose need not be administered as a single injection but may comprise continuous infusion over a set period of time.
  • a unit dose may contain at least 0.01, 0.05, 0.1, 0.5, 1.0, 2.5, 5.0, 10.0, 15.0, 25.0 or 50.0 mg of the active ingredient.
  • a unit dose contains less than 0.01, 0.05, 0.1, 0.5, 1.0, 2.5, 5.0, 10.0, 15.0, 25.0 or 50.0 mg of the active ingredient.
  • a unit dose contains from about 0.001 mg to about 50 mg of the active ingredient.
  • a unit dose contains from about 1 mg to about 10 mg of active ingredient.
  • the vaccine composition may be administered in a single daily dose, or the total daily dosage may be administered in divided doses, for example, two, three or four times daily.
  • the vaccine composition may be administered in intranasal form via topical use of suitable intranasal vehicles, via transdermal routes, using those forms of transdermal skin patches well known to persons skilled in the art, by implantable pumps; or by any other suitable means of administration.
  • the dosage administration will, of course, be continuous rather than intermittent throughout the dosage regimen.
  • the dosage regimen utilizing the vaccine composition is selected in accordance with a variety of factors including type, species, age, weight, sex and medical condition of the patient; the severity of the condition to be treated; the route of administration; the renal and hepatic function of the patient; and the particular compound employed.
  • a physician or veterinarian of ordinary skill can readily determine and prescribe the effective amount of the drug required to prevent, counter or arrest the progress of the disease or disorder that is being treated.
  • Vaccine administration may further comprise a prime-boost regimen.
  • a prime-boost regimen In these methods, one or more priming immunisations are followed by one or more boosting immunisations.
  • the actual immunogenic composition can be the same or different for each immunisation and the type of immunogenic composition, the route, and formulation of the immunogens can also be varied.
  • One useful prime-boost regimen provides for two priming immunisations, four weeks apart, followed by two boosting immunisations at 4 and 8 weeks after the last priming immunisation.
  • Such a regimen may comprise, for example, priming with full-length gp41 and boosting with one or more immunogenic peptides as disclosed herein.
  • Either the prime or the bosst, or both, may be administered in the form of a DNA molecule encoding the peptide or polypeptide in question.
  • this regimen might elicit gp41 cross-reactive antibodies targeted to the epitopes present in the priming immunogen.
  • the designed protein fragments may be expressed in E. coli in order to prevent glycosylation and consequent epitope masking that might occur if expressed in a eukaryotic expression system.
  • the use of E. coli to produce non-glycosylated versions of the invention may have contributed to the success of the approach.
  • Immunisation schedules are well known for animals (including humans) and can be readily determined for the particular subject and immunogenic composition.
  • the immunogens can be administered one or more times to the subject.
  • there is a set time interval between separate administrations of the immunogenic composition typically it ranges from 10 days to several weeks, and is often 2, 4, 6 or 8 weeks.
  • the interval is typically from 2 to 6 weeks.
  • the interval is longer, advantageously about 10 weeks, 12 weeks, 14 weeks, 16 weeks, 18 weeks, 20 weeks, 22 weeks, 24 weeks, 26 weeks, 28 weeks, 30 weeks, 32 weeks, 34 weeks, 36 weeks, 38 weeks, 40 weeks, 42 weeks, 44 weeks, 46 weeks, 48 weeks, 50 weeks, 52 weeks, 54 weeks, 56 weeks, 58 weeks, 60 weeks, 62 weeks, 64 weeks, 66 weeks, 68 weeks or 70 weeks.
  • the immunisation regimes typically have from 1 to 6 administrations of the immunogenic composition, but may have as few as 1, 2, 3, 4 or 5.
  • the methods of inducing an immune response can also include administration of an adjuvant with the immunogens.
  • annual, biannual or other long interval (5-10 years) booster immunisation can supplement the initial immunisation protocol.
  • a specific embodiment of the invention provides methods of inducing an immune response against HIV in a subject by administering an immunogenic composition of the invention, , one or more times to a subject wherein the peptic sequence is expressed at a level sufficient to induce a specific immune response in the subject.
  • Such immunisations can be repeated multiple times at time intervals of at least 2, 4 or 6 weeks (or more) in accordance with a desired immunisation regime.
  • the immunogenic compositions of the invention can be administered alone, or can be co-administered, or sequentially administered, with other HIV immunogens and/or HIV immunogenic compositions, e.g., with ‘other’ immunological, antigenic or vaccine or therapeutic compositions thereby providing multivalent or ‘cocktail’ or combination compositions of the invention and methods of employing them.
  • the ingredients and manner (sequential or co-administration) of administration, as well as dosages can be determined taking into consideration such factors as the age, sex, weight, species and condition of the particular subject, and the route of administration.
  • the other HIV immunogens can be administered at the same time or at different times as part of an overall immunisation regime, e.g., as part of a prime-boost regimen or other immunisation protocol.
  • HIV immunogens are known in the art.
  • HIVA (described in WO 01/47955), which can be administered as a protein, on a plasmid (e.g., pTHr.HIVA) or in a viral vector (e.g., MVA.HIVA).
  • RENTA (described in PCT/US2004/037699), which can also be administered as a protein, on a plasmid (e.g., pTHr.RENTA) or in a viral vector (e.g., MVA.RENTA).
  • one method of inducing an immune response against HIV in a human subject comprises administering at least one priming dose of an HIV immunogen and at least one boosting dose of an HIV immunogen, wherein the immunogen in each dose can be the same or different, provided that at least one of the immunogens is an epitope of the present invention, a nucleic acid encoding an epitope of the invention or an expression vector, preferably a VSV vector, encoding an epitope of the invention, and wherein the immunogens are administered in an amount or expressed at a level sufficient to induce an HIV-specific immune response in the subject.
  • the HIV-specific immune response can include an HIV-specific T-cell immune response or an HIV-specific B-cell immune response.
  • Such immunisations can be done at intervals, preferably of at least 2-6 or more weeks.
  • Treatment includes both therapeutic treatment and prophylactic or preventative treatment, wherein the object is to prevent or slow down the targeted pathologic condition or disorder.
  • Those in need of treatment include those already with the disorder as well as those prone to have the disorder or those in whom the disorder is to be prevented.
  • the terms ‘therapy’, ’therapeutic’, ‘treatment’ or ‘treating’ include reducing, alleviating or inhibiting or eliminating the symptoms or progress of a disease, as well as treatment intended to reduce, alleviate, inhibit or eliminate said symptoms or progress.
  • Desirable effects of treatment include preventing occurrence or recurrence of disease, alleviation of symptoms, diminishment of any direct or indirect pathological consequences of the disease, preventing metastasis, decreasing the rate of disease progression, amelioration or palliation of the disease state, and remission or improved prognosis.
  • methods and compositions of the invention are used to delay development of a disease or disorder or to slow the progression of a disease or disorder.
  • an effective amount preferably a therapeutically effective amount of the (poly)peptide or vector of the invention is administered.
  • An ‘effective amount’ refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired therapeutic or prophylactic result.
  • the effective amount may vary according to the drug or prodrug with which the (poly) peptide or vector is co-administered.
  • a ‘therapeutically effective amount’ of a (poly) peptide or vector of the invention may vary according to factors such as the disease state, age, sex, and weight of the individual, and the ability of the (poly) peptide, to elicit a desired therapeutic result.
  • a therapeutically effective amount encompasses an amount in which any toxic or detrimental effects of the (poly) peptide are outweighed by the therapeutically beneficial effects.
  • a therapeutically effective amount also encompasses an amount sufficient to confer benefit, e.g., clinical benefit.
  • isolated peptide or nucleic acid may be isolated substantially or completely away from one or more elements with which is associated in nature, such as other naturally occurring peptide or nucleic acids or other peptide or nucleic acid sequences.
  • FIGURES are a diagrammatic representation of FIGURES.
  • FIGURE 1 FabA 43 conformational epitope P7 designed in silico blocks FabA 43 binding to gp41 and reverse FabA 43 neutralization activity.
  • A Preincubation of FabA 43 with the conformational epitope P7 (5 mM, hatched bars) but not with control HA (5mM, plain bars) interferes significantly with FabA 43 binding to clade A, clade B and clade C PI or clades A and C gpl40 and clade B gp41, as evaluated by ELISA.
  • B The P7-induced reduction of FabA 43 binding to each antigen is dose dependent.
  • Binding inhibition to FabA 43 binding to each antigen in the presence of P7 is calculated relative to the binding inhibition in the presence of the irrelevant HA peptide serving as negative control.
  • values represent mean ⁇ SEM, derived from 3 independent experiments performed in triplicate.
  • C Localization of the amino acids paths corresponding to P7 peptide on the 6-Helix bundle conformation of clade A, B and C gp41 framel5. Each gp41 monomer of the trimer is depicted using a different tone of gray. The amino acid paths corresponding to the FabA43 specific peptide P7 are highlighted in orange.
  • residues 532-535 belong to the N-Helix of one monomer while residues 669- 672 belong to the C-Helix of another monomer of the trimer.
  • Red the P7 predicted conformation superimposed on the structure of gp41.
  • RMSD values over the paired residues of gp41 and P7 are of 2.45, 2.97 and 1.97 A for clade A, B and C, respectively. Note that in this region of the gp41, only residue 535 differs between clade B in one hand and clade A and C in the other hand.
  • D Conformational variability of the predicted conformations of P7 using PEP-FOLD3. Left: conformation best fitting gp41. Right: top 10 predicted conformations superimposed.
  • FIGURE 2 P7 neutralisation of HIV infection of CD4+T cells
  • P7 induce inhibition of CD4 + T cells infection in a dose-dependent manner.
  • the potential of P7 to inhibit CD4 + T cells infection was assessed in a single cycle infectivity assay, using p24 staining on primary CD4 + T cells.
  • P7 at indicated concentrations, was preincubated for 1 h at 37° with either free virus (dotted line, diamond) or activated CD4 + T cells (dotted line, triangle). After lh, activated CD4 + T cells were added when P7 was preincubated with the virus, or virus was added when P7 was preincubated with CD4 + T cells. Cultures were then incubated for 36 h.
  • the percentage of neutralization was defined as the reduction of the Gag-p24 + CD4 + T cells in the presence of P7 compared with HIV- 1 -infected CD4 + T cells in the absence of P7 (solid line, circle). Values represent mean ⁇ SEM, derived from at least 4 independent experiments performed in triplicate.
  • FIGURE 3 P7 binds to trimeric envelope gpl40 cross clade
  • the gp41 (584-684) used was a construct based on the HXB2 group M subtype B sequence [40] and kindly provided by Dr. Weissenhorn. Gpl40 from HIV-1 clades A (92RW020) and C (C.ZA.1197MB) were obtained from Immune Technology Corp (NY10021, USA).
  • PI a.a 630-685 derived from clade B HXB2 gp41 (Pl-B) was SQTQQEKNEQELEELDKWASLWNWFDITNWLWYIK (SEQ IDN°2) as described [22], from clade A 99UGA07072 gp41 (PI -A) was SQIQQKKNEQDLLALDKWANLWNWFDISNWLWYIR (SEQ IDN°3) and from Clade C Bw96Bw0502 (Pl-C) was SQTQQEKNEQELLALDS WKNLWNWF SITNWLWYIK (SEQ IDN°4).
  • Peptides were chemically synthesized at a purity >95% by Biopeptide (LA, USA) for Pl-B and United BioSystems (VA, USA) for Pl-A and -C.
  • ESN Fab As were transformed in their corresponding FabGs by molecular cloning using pASK88 vector which direct the synthesis of the human gamma- 1 heavy chain and light chain, respectively [41] Production of functional Fabs was performed as previously described [2] Briefly, cultures were grown in 1L of LB medium containing 100pg/ml ampicillin and expression was induced for 14 hr at 22°C by addition of 0.2 mg/L of anhydrotetracycline (ACROS Chimica) at an OD550 of 0.5. Fabs were purified from the periplasmic fraction of the E.
  • ACROS Chimica anhydrotetracycline
  • the concentration of FabA and G was measured by sandwich ELISA allowing the restricted detection of full Fabs, namely covalently linked heterodimers, using goat anti human IgA or IgG (Caltag, France) for coating, and biotinylated mouse anti-human Ig kappa light chains (B.D. Pharmingen, USA) for detection, as earlier described [2]
  • SPR Surface Plasmon Resonance
  • the final immobilization levels were between 300 and 500 RU to avoid rebinding events, as mentioned in [42]
  • Initial binding experiments with clone 43 and 177 of FabA and G, and non-specific isotypes as analytes were comparatively performed. No specific signal was observed with non-specific isotypes (recorded value of 0 RU), indicating that the observed binding of Fabs the different HIV-1 envelope subunits was specific.
  • sensorgrams were obtained by passing various concentrations of the analytes at various concentrations as indicated, over the ligand surface, at a flow rate of 50pl/min, with a 3-min association phase and a 6-min dissociation phase.
  • the sensor surface was regenerated between each experiment with a single injection of 35 mM NaOH and 1.3 M NaCl at a flow rate of 50 m ⁇ /min for 30sec. Identical injections over blank surfaces ran in parallel (and giving a value of 0 RU) were subtracted from the data for kinetic analysis. Binding kinetics were evaluated by linearization using “fit kinetics Langmuir binding type” with BiaEvaluation software (BiacoreTM). Relative Pearson's chi- squared tests assessing goodness of fit were always below 10, which indicated that the models used for fitting adequately describe recorded data.
  • ELISA binding assays were performed as described [11] by coating microtiter plates (NUNC-Immuno Plate MaxiSorp Surface, or Peptide Immobilizer Exiqon Peptide Immobilizer, Exiqon) with gpl40 (trimeric rgp41 at 0.25 microg/well), PI (0.1 pg/well), peptides corresponding to conformational epitopes (0.1 pg/well), overnight at 4°C in PBS.
  • Fab binding was detected with a biotinylated mouse anti-human Ig kappa light chains (BD Pharmingen) and streptavidin- HRP or with anti-human IgA HRP (Jackson ImmunoRe search) for the peptides corresponding to conformational epitopes. All experiments were performed with Fabs from at least three independent purifications, each in duplicate.
  • Fab 43 was preincubated ON at 4°C with various concentration of P7 or the irrelevant Hemagglutinin (HA) (YPYDVPDY) peptide serving as negative control and added to gp41 clade B, gpl40 (clades A, C) or PI (clades A, B, C) coated on the ELISA plate at a final concentration of 0.8nM for FabA 43 and 0.05 to 1 OmM for P7.
  • Fab binding to each gp41 subunit was finally detected enzymatically by using anti human IgA coupled with HRP.
  • Fab binding to HIV-1 infected cells Binding assays were performed as described [43] For detection of Fab binding to native HIV-1 envelope at the surface of HIV-1 infected cells, FabA or G (5pg/ml) were incubated with 105 cells overnight at 4°C. To allow for direct isotype comparison, FabA and G were detected in parallel using the same fluorescein isothiocyanate (FITC)-conjugated mouse anti-human kappa light chain (BD Biosciences, San Jose CA, USA), 30 minutes at 4°C.
  • FITC fluorescein isothiocyanate
  • Peripheral blood samples from healthy donors obtained from the Etableau Franqais de Sang (Paris, France) were depleted of CD8+T cells with Rosette Sep cocktail, (StemCell Technologies Inc., France) and peripheral blood mononuclear cells (PBMC) were isolated by Ficoll-Hypaque. After stimulation for 2 days with 5pg/ml phytohemagglutinin (Sigma-Aldrich, St.Louis, MO) as described [11], CD8-depleted PBMCs were used for infection and neutralization experiments. Alternatively, CD4+T CEM-NKR lymphocytic cells (NK-resistant) expressing CCR5 (AIDS Research and Reference Program, NIH) we used as when indicated. To prepare CD4+ T target cells for neutralization assays, cells were split 1 :3 on the day of passage and used the following day.
  • a stock of HIV-1JR-CSF (clade B, R5 tropic) was prepared on a large scale by transfecting 293T cells with a plasmid containing the DNA sequence of JR-CSF (NIH, Germantown, MD USA) [2] The cell culture supernatant was concentrated, separated into single use aliquots and stored at -80 deg C.
  • Single-cycle neutralization assay The neutralization activity of FabA and FabG was evaluated on primary CD4+ T-cells (CD8+T cells-depleted PBMC), on CEM-CCR5+ infected with HIV-1 JR-CSF (Clade B) or with each of Clade A or C primary isolates and quantified by flow cytometry after intracellular Gag-p24 staining, as we described earlier [2] At least five independent experiments, performed each in triplicate, were performed. Live cells initially gated by forward and side scatter were analyzed for intracellular expression of p24-Ag. A dose dependent parameter was used to compare the FabA with FabG and for determination of maximum percent inhibition values. Neutralization was defined in % of cells infected in the absence of antibody. Titers were calculated as IC50 and IC85 and reported as the concentration of Fab causing a 50 or 80% reduction in the percentage of p24+ cells compared to virus control wells.
  • FabA 43 was preincubated with a 400-fold molar excess of P7 overnight at 4°C and further incubated the virus and the target primary CD4+ T-cells at final FabA 43 concentration of 20nM and P7 concentration of 33 mM.
  • LCs monocytes derived Langerhans cells
  • TCD4+ cells autologous TCD4+ cells
  • the virus transfer was evaluated by measuring Gag-p24 released in the culture medium using a commercial ELISA (Innotest HIV-1 Antigen mAb, Innogenetics) according to manufacturer instructions. Results are expressed as % of inhibition transfer using formula [(LC+T-Ab) - (LC+T+Ab) / (LC+T-Ab)] xlOO.
  • Epitope mapping of both isotypes was performed as we described previously [11], using linear 12-mer peptide libraries displayed on the protein pill of M13 phages (New England Biolabs) as recommended by the manufacturer.
  • IgA- or IgG-coated beads were incubated with Fab IgA or IgG on a rotating wheel for 2 h at room temperature and epitope screening was initiated by incubating each bead of Fab IgA or IgG with the original 12-mer (1013) phage displayed peptide library containing different phages, overnight at 4°C.
  • the crystal structure of the 6-Helix bundle HIV-1 gp41 including both fusion peptide and membrane proximal external regions [33] was retrieved from the RCSB Protein Data Bank (http://www.rcsb.org, code : 2X7R).
  • the solution NMR structure of ectodomain of SIV gp41 [48] was retrieved from the RCSB Protein Data Bank (http://www.rcsb.org, code : 2EZO). Multiple sequence alignment was used to build the 6- Helix bundle structure of HIV- 1 gp41.
  • Pre fusion clade B gp41 The structure of the trimeric gp41 ectodomain of HIV- 1 in its pre-fusion state was built using the sole crystallographic structure of the pre-fusion HIV-1 gp41 and missing loops were modeled using the BCLoopSearch algorithm (http://bioserv.rpbs.univ-paris-diderot.fr/services/BCSearch/) which helps identify linear fragments similar to a query in large collections of structures using a new similarity approach based on a Binet Cauchy (BC) kernel [49]
  • BCLoopSearch algorithm http://bioserv.rpbs.univ-paris-diderot.fr/services/BCSearch/
  • the crystal structure of the pre-fusion HIV-1 gp41 [32] was retrieved from the RCSB Protein Data Bank (http://www.rcsb.org, code : 4TVP).
  • Pre and post fusion Clade A and C gp41 The clade A and clade C gp41 sequences were retrieved from http://www.hiv.lanl.gov (code : AF484478 and code : AF110967, respectively) aligned and mapped onto the previous structures using the MODELLER v9.15 software [46]
  • the box was then filled with water and we added a concentration of 0,2 M Na+Cl- to the solvent, as well as an excess of Na+ ions to neutralize the system charge induced by the negatively charged residues. 200 ns of Molecular Dynamics simulations were then run for the structures using Gromacs 2016.1 [50] [51] with AMBER99SB-ILDN [52] as force field and TIP3P [53] model for water.
  • a discontinuous 3D epitope is localized on the protein surface by searching for a 3D fit with partial amino acid strings of a given sequence in a preset distance.
  • PepSurf compares each peptide provided to the solvent accessible surface of the antigen, determining the best path.
  • the algorithm then creates clusters of antigen residues on the surface that best fit a grouping of peptides and returns a score for each residue at the surface corresponding to the number of occurrences of this residue in the alignments.
  • Snapshots of the gp41 trimeric structures were taken every 10 ns of the MD starting from 60 ns and explored using the pepSurf algorithm with both BLOSUM62 and BLOSUM80 as similarity matrix and the 141 antibody binding mimotopes as input.
  • a library of 147 12-mer soluble peptides was generated using SolyPep on the RPBS web portal and used also as input to account for the background noise. Scores were then averaged for each residue position for the three chains of the 15 structure snapshots to account for the protein flexibility. Scores obtained for the random peptides library were subtracted.
  • Conformational paths - that is, paths that encompass two different chains - were selected and the resulting sequence modeled with the PEP-FOLD3 algorithm. Only peptide structures that aligned to the same sequence in the gp41 structure with a RMSD lower than 2,5 angstroms were retained, in order to keep only the peptide that would fold in water in the same manner as in the gp41 structures.
  • FabA 43 has a high degree of somatic mutations, a VH3 heavy chain origin similar to the broadly neutralizing IgG 4E10 [20], and a long CDRH3 of 22 residues, features characteristics of other broadly neutralizing IgGs [2]
  • FabA 177 has a low level of somatic mutations for their heavy chain, 100% homology with germ-line gene region IGKVID-30*-01, and a normal length CDRH3 of 11 residues [2]
  • FabA 177 heavy chain origin is VH6 family, which was never described before for broadly neutralizing IgGs.
  • Each FabA was transformed by genetic engineering replacing the CHlal into CHlyl , as described in the method section, in their corresponding FabG, the two isotypes thus sharing identical VH and VL domains and the same light chain, but expressing different CHI domain.
  • FabA 43 and 177 recognize conformational conserved regions on clade B gp41 from both R5 and X4 tropic viruses [2]
  • both FabA and G pairs specifically bind to Clade B gp41 as well as to Clade A and C gpl40 in a dose-dependent manner, but strikingly for all clades, binding of FabA 43 and 177 is more efficient compared to their respective FabG Hence, a 50 times higher concentration of FabG 43 (50pg/ml) is required to reach the same binding of FabA 43 (lpg/ml)).
  • SPR Surface plasmon resonance
  • KDs are 6.41 and 4.79nM for the FabA 43 compared to 0.50mM and no affinity is measured for its respective FabG; KD equals 2.92 and 2.15nM for FabA 177 whereas its corresponding FabG has no measurable affinity for any of the clade A or C viral envelope.
  • affinities reach the nM range for the FabA, whereas remain in the order of mM or even lower for the FabG for both clones, in agreement with binding experiments using ELISA and show that the affinity of both FabA 43 and 177 are greater, for all clades, than that of their FabG counterpart.
  • FabA 43 and 177 bind to clade B gp41 and clade A and C gpl40 with nM affinity whereas affinities of corresponding FabG are much lower, although still able to bind their targets as shown by ELISA approaches.
  • the 35 amino acid peptide PI was characterized as the minimal membrane proximal external region of the viral envelope gp41 that allows for HIV-1 binding to galactosyl ceramide, the HIV-1 mucosal receptor and for mucosal HIV-1 entry by transcytosis [21] [22] [23] This highly conserved region has been used as immunogen in a prophylactic vaccine against HIV-1 both in pre-clinical and clinical phase I trials [19, 24] PI sequence is well conserved between clade B and A viruses whereas a K670S mutation in clade C virus prevents binding of the broadly neutralizing 2F5 IgG [25]
  • FabA 43 was obtained by screening the FabA ESN library on clade B PI and consequently FabA 43 binds to clade B PI [2] We thus evaluated by ELISA the capacity of FabA and G 43 to bind to clade A, B and C PI, comparatively as described [2] Both Fab 43 isotypes bind
  • FabA 43 recognizes PI more efficiently than its corresponding FabG. This difference is higher when clade B PI is the target (with a 60- fold change from FabA to G), but still significant (with a 20-fold change from FabA to G) when Clade A and C are targets. Additionally, FabA and G 43 bind more efficiently to Clade B compared to Clade A and C PI. These differences in Fab isotype binding to PI from the three clades are also observed by SPR.
  • FabA 43 and 177 affinities for the gp41 viral envelope cross clade is in the nM range, similar to that of other broadly neutralizing Abs (bNAbs) [26] whereas FabG recognize less efficiently the same antigen.
  • FabA 43 and 177 bind HIV-1 infected cells better than their respective FabG with 20-40% of T-cells labelled for FabA compared to 5-19% for FabG (Student’s t -test, p ⁇ 0.05) for Fab 43 and 177 respectively).
  • FabA 43 recognizes more efficiently Clade A than clade B and C infected CD4 + T-cells (Student’s t -test, p ⁇ 0.05), whereas no differences were apparent between clades for FabA 177.
  • CHI domain can affect the paratope and its fitting on the antigen and may in turn impact the Ab anti-viral function in an isotype-dependent manner.
  • the CHla provides FabA with the capacity to neutralize CD4 + T-cell infection whereas the corresponding FabG harboring the same paratope but a CHly has limited neutralizing activities.
  • HIV-1 entry through multilayer mucosal tissues occurs mainly by targeting Langerhans cells (LCs) that in turn transfer to CD4 + T-cells [28] [29] [30] [31] that constitute the founder infected population.
  • LCs Langerhans cells
  • CD4 + T cells can disseminate out of the mucosal tissue and spread the virus to other CD4 + T cells.
  • Transfer of clade A and C HIV-1 is inhibited by both FabA 43 and 177 in a concentration dependent manner, as well as that of clade B by FabA 43, inducing a >40% inhibition at lOOng/ml. Transfer of clade B virus remains much less sensitive to FabA 177 .
  • FabG 43 blocks transfer of Clade A and C viruses but less efficiently than its FabA counterpart and FabG 177 was only able to block clade A HIV-1 transfer.
  • a 12mer random peptide library was used to characterize a set of the best specific peptides, referred to as mimotopes, for both 43 and 177 clones as FabA and FabG using three rounds of successive screening with increasing stringency, as we described [2]
  • each set of FabA mimotopes was docked onto trimeric gp41 crystal structures using in silico approaches, as described in the method section and supplementary data.
  • Only two gp41 crystal structures of a Clade B gp41 in different conformational states are available.
  • One structure represents gp41 in the pre-fusion state together with gpl20, although this gp41 lacks the MPER region [32]
  • the other gp41 structure mimics the post fusion state and is composed of three N-helices and three C-helices that form a six helix-bundle for completion of HIV- 1 fusion with target cells in a spring load model of fusion.
  • the gp41 construct lacks the gp41 Cys- loop bridging the C and N helices [33]
  • Clone 43 is primarily specific for the gp41 MPER [2] and the Cys-loop contains important gp41 epitopes [34] Therefore, we first optimized the available crystal structure of gp41 clade B by reconstituting the missing MPER in the pre-fusion structure and the missing loop in the 6-Helix bundle as described in the supplementary data. As no crystal structure of clade A or C are accessible in the literature, we constructed A and clade C gp41 structure by mapping each of clade A and Clade C gp41 sequences onto the clade B gp41 structures, as detailed in the supplementary data. As a result, from these in silico modeling calculations, Clade A, B and C gp41 structures in the pre- and 6-Helix bundle conformation were available for epitope docking studies.
  • FabA 43 mimotopes Selected FabA specific mimotopes obtained from FabA 43 and 177 were localized on clade A, B and C gp41 pre-fusion and six-helix bundle structures using the Pepsurf method as detailed in supplementary data. Three main regions were targeted by FabA 43 mimotopes on gp41 helix bundle structure of all three clades the highest hits being localized on the lower MPER region interface with the N-helix, in agreement with the screening strategy focusing on PI used for selecting FabA 43 [2] In comparison, FabA 43 mimotopes also fit on the pre-fusion structures, although in a more scattered manner and with lower scores.
  • the main regions targeted by FabA 177 mimotopes on gp41 helix bundle structure of all three clades differ, as expected from the FabA 177 screening strategy, from those targeted by FabA 43 mimotopes, and the best fit appears in the loop linking the N and C gp41-helices.
  • These FabA 177 mimotopes also match gp41 pre-fusion structures, but with lower scores and in a more scattered pattern, irrespective of the clade.
  • P7 One out of 5 conformational epitopes defined in silico for FabA 43, referred to as P7, competed significantly with FabA 43 binding to PI and gp41/gpl40 of the three clades in ELISA when preincubated with the FabA 43 ( Figure 1A) in a concentration-dependent manner (Figure IB). P7 at 5 mM could block FabA 43 binding by >50%, reaching >80% at 1 OmM ( Figure IB). A 9 aa peptide derived from the influenza virus hemagglutinin (HA) used as negative control did not interfere with 177 binding in ELISA ( Figure 1 A).
  • HA hemagglutinin
  • P7 did not interfere with Fab G 43 binding to the same antigens (not shown).
  • P7 interfere with FabA 43 cross-clade.
  • P7 corresponds to an 11 aminoacid peptide located at the interface formed by the tips of Isl and H-helices of the three clades of gp41 ( Figure 1C). This cross-clade activity is unexpected as P7 has been defined as the best conformational epitope extracted from in silico on FabA 43 mimotope fitting to the 6-Helix bundle Clade A gp41 structure.
  • the other four conformational peptides defined in silico for FabA 43 that do not interfere with Fab A 43 binding to the gp41 antigens are located on different regions of gp41 on each gp41 clade, although for two of them spatially close from the region defined by.
  • Lamiable, A., et al., PEP-FOLD3 faster de novo structure prediction for linear peptides in solution and in complex. Nucleic Acids Res, 2016. 44(W1): p. W449-54.

Abstract

The inventors used a reverse vaccinology approach based on the Fab conformational epitopes to generate a (poly) peptide that is useful in therapeutic approaches against HIV infection. Accordingly, the invention relates to a novel peptide relating to HIV is disclosed and therapeutic compositions containing such peptide are discloseda. The compound and compositions of the invention are of use in prevention and treatment of HIV, and in particular for inducing mucosal immunity to HIV.

Description

VACCINE COMPOSITIONS FOR HIV PREVENTION AND TREATMENT
FIELD OF THE INVENTION
The invention is in the infection field. More particularly, the invention relates to novel (poly) peptides relating to HIV are disclosed, as are vaccine compositions containing such (poly) peptides. The compounds and compositions of the invention are of use in prevention and treatment of HIV, and in particular for inducing mucosal immunity to HIV. BACKGROUND OF THE INVENTION
HIV-1 infection is mainly initiated at mucosal sites during sexual intercourses, and IgA can efficiently prevent HIV-1 infection and mucosal reservoir establishment. The inventor and others have previously shown that mucosal IgAs, specific for HIV-1 envelope gp41-subunit conserved epitopes, naturally raise in 1% of individuals upon sexual exposure to HIV-1 who remain persistently uninfected. These individuals, referred to as Exposed SeroNegatives (ESN), remain IgG seronegative but raise gp41 -specific mucosal IgA that constitute a main correlate of protection (reviewed in [1] [2] [5]). In vitro, ESN IgA are able to inhibit HIV-1 mucosal penetration by transcytosis across epithelial barriers and to neutralize CD4+T-cell infection against Clade B HIV-1 [2] [6] [7] These IgA, detected mainly in vaginal washes and urine, recognize the conserved regions of the transmembrane subunit gp41 of HIV-1 envelope [6] [2, 7] Remarkably, at the molecular level, ESN IgAs are affinity matured with extensive somatic hypermutations, and have extended heavy-chain third complementary determining regions, (CDRH3) [2], characteristics similar to those found for IgGs in natural HIV-1 infection and that increase breadth and potency in vitro [8] [9] [10] Compared to HIV-1 infected patient IgGs, which unfortunately develop too late (several years) to prevent infection, mucosal ESN IgAs raise early after HIV-1 exposure and offer protection from infection. The presence of ESN protective gp41 -specific IgAs at mucosal level evidences an efficient “natural” vaccination. Furthermore, this IgA-driven immune response in ESN questions about the protective mechanisms of a humoral response involving only mucosal IgA, and, at the molecular level, about the participation of the isotype specific constant regions of the antibody in ESN IgA efficacy.
Antibodies (Ab) have been historically described as being composed of two distinct structurally independent domains, namely the variable regions, forming the paratope, responsible for antigen binding, and the heavy chain constant regions mediating effector functions cells such as antibody dependent cell cytotoxicity (ADCC) and antibody dependent phagocytosis (ADCP). Recent works including those of the inventor [11], revealed that the Ab isotype carried by the Ab heavy chain constant region (CH) influences the paratope structure and in turn the antigen recognition. Modulation of antigen recognition by CH has been reported for IgM, IgG, IgE and IgA, using a variety of biochemical and biophysical techniques. Hence, by isotype switching a broadly neutralizing 2F5 IgGl specific for gp41 derived from the blood of an HIV- 1 -infected patient [13] into a IgA2, the inventor previously showed that the CH influences affinity and epitope specificity, and in turn, antiviral functions [11] Changes induced by isotype switching occur most likely by modulation of the paratope conformation due to non-local [14] and even allosteric effects that modify Ab- antigen interaction [15], antigen binding affecting the constant region and vice versa, as hypothesized by Ofran and col. [16] The CH contains three domains, namely CHI, CH2 and CH3. CH3 and CH2 participate in Fc- receptor binding on effector cells to mediate ADCC or ADCP whereas the CHI domain, separated from the two others by the hinge region, is part of the Fab region of the Ab and closer to the paratope formed by the variable heavy (VH) and light (VL) chains. Which CH domain(s) participates in modulation of Ab specificity and affinity remains unclear and whether the CHI itself, due to its proximity to the VH and VL regions, plays a determinant role in Ab affinity and functions has not been studied. Furthermore, the role of constant regions has only been evaluated by comparing blood derived antibodies expressing identical V regions in different heavy chain constant context and thus addressing a blood raised B-cell epitope. However, mucosal immunity is highly compartmentalized [17] resulting in the induction of mucosal antibody recognizing B cell epitope that differ from systemic one. Whether the antibody isoptype also affects Ab specificity and affinity and even function of mucosal Abs in a similar manner than blood derived Abs remains unknown.
Most of the vaccine approaches against HIV-I have targeted the viral envelope (Env) glycoproteins because they are the major surface antigens expressed on virions and by HIV-I infected cells. The native HIV-I envelope glycoprotein is a heterotrimer containing three gpl20 proteins non-covalently associated with three gp41 glycoproteins.
The three most potent HIV-I neutralizing antibodies yet identified, bl2, 2G12 and 2F5, have a high affinity for the native trimer. There is increasing effort to develop recombinant proteins as candidate vaccines that are better antigenic mimics than the native envelope glycoprotein complex such as the gpl20/gp41. However, due to numerous molecular homologies between the gpl20/gp41 and molecules of the immune system, many potential cross-reactivities events may occur leading to possible harmful autoimmune phenomena.
Various strategies have been described in order to dampen those cross- reactivities for obtaining anti-HIV vaccines with no or less cross reactivities with human proteins as the introduction of mutations and/or deletions in various part of the gp41 proteins as described in US 6,455,265 and WO 2005/010033.
However there is still a need for a vaccine that allows for inducing a versatile immune response against HIV infection, and in particular HIV-type 1 infection. There is also a need for the development of non-clade B vaccines, such as, for example, clade A and C strains.
There is also a need for the development of a vaccine with broad inhibitory spectrum allowing for cross-clade inhibition.
There is a need for a vaccine allowing to induce a humoral and/or cellular immune response against HIV infection.
There is a need for a vaccine allowing to induce an immune response against HIV infection at the mucosal surface level and at the blood level.
There is a need for a vaccine suitable for inducing mucosal IgA antibodies and systemic IgG antibodies.
It is an object of the invention to satisfy to all those above-mentioned needs. DESCRIPTION OF THE INVENTION
The inventor took advantage of two ESN mucosal anti-HIV-1 Fab-IgAl (FabA) previously characterized [2], that were isotype switched into Fab-IgGl (FabG). Each FabA and its respective isotype switched FabG were compared for HIV-1 envelope binding, epitope recognition, and anti- viral activities against the main HIV-1 strains present worldwide, namely HIV-1 of Clades B, A and C, and their conformational epitopes on gp41 were determined using in silico analyses. These comparative analyses result in a molecular understanding of the recognition of different epitopes on HIV-1 envelope, depending on the isotype in correlation with different affinities and antiviral functions. Based on these results, the inventors used a reverse vaccinology approach based on the Fab conformational epitopes to generate a (poly) peptide that is useful in therapeutic approaches against HIV infection.
Thus, in one aspect the invention provides a peptide comprising or consisting of the consecutive sequence of amino acids (SEQ IDN°1): LWNWFDISAASI
The peptide is useful in preventive and curative treatments of HIV infections.
In a first aspect, the invention provides a therapeutic composition comprising a peptide of the invention, wherein the peptide is used for its direct neutralizing properties against an HIV infection, namely by clade A, B and C virus.
According to this aspect, the peptide is used preferably systemically, as an antiviral agent.
In a second aspect, the invention provides a therapeutic composition comprising a peptide of the invention, wherein the peptide is used for its antigenic properties for inducing a protective immunity against an HIV infection.
Also provided are nucleic acids encoding the peptides of the invention, vectors comprising said nucleic acids and host cells comprising said vectors and nucleic acids.
Also provided is a composition comprising a peptide, or nucleic acid of the invention. In a preferred embodiment, said composition is a vaccine composition. In some embodiments, said composition is suitable for mucosal administration. For example, said composition may be suitable for administration to the nasal, rectal or vaginal mucosa. In some embodiments, said vaccine composition additionally comprises one or more adjuvants.
Also provided is a peptide, nucleic acid or composition of the invention for use in the preventive or therapeutic treatment of HIV or AIDS. The composition may be for administration to a mucosal surface, or any other administrative route such as those described below. The composition may be for administration in a prime-boost regimen such as those described below, such as a regimen comprising administration of full-length HIV-1 gp41.
Also provided is a peptide, nucleic acid or composition of the invention for use in a method of inducing a mucosal immune response to HIV-1 n some embodiments, said method comprises administering said peptide, nucleic acid or composition to a subject, in particular a subject in need thereof. The peptide, nucleic acid or composition may be administered to a mucosal surface of a subject, for example the nasal, rectal or vaginal mucosa. Various administration regimens are described herein and encompassed within the scope of the invention.
Also provided is a peptide, nucleic acid or composition of the invention for use in a method of inducing a mixed mucosal IgGl and IgA2 response to HIV-1, the method comprising administration of at least a peptide comprising the peptidic sequence of the invention or nucleic acids encoding said peptides, or a composition comprising said peptides or nucleic acids.
Also provided is the use of a peptide, nucleic acid or composition of the invention for use in the preparation of a medicament for the preventive treatment of HIV infection, in particular HIV- 1 infection, or AIDS.
Also provided is a method of prevention or treatment of HIV infection, in particular HIV-1 infection, comprising administration of a composition of the invention to an individual in need thereof. In some embodiments, said composition is administered to a mucosal surface of a subject, for example the nasal, rectal or vaginal mucosa. Various administration regimens are described herein and encompassed within the scope of the invention.
Also provided is a method of inducing a mucosal immune response to HIV-1, comprising prevention or treatment of HIV infection, comprising administration of a composition of the invention to an individual in need thereof. In some embodiments, said composition is administered to a mucosal surface of a subject, for example the nasal, rectal or vaginal mucosa. Various administration regimens are described herein.
Also provided is a method of inducing a mucosal IgA response to HIV-1, the method comprising administering at least one peptide as defined above or a nucleic acid encoding said peptide, or a composition comprising said peptide or nucleic acid, to an individual in need thereof, as described herein.
Also provided is a method of inducing a mucosal IgG response to HIV-1, the method comprising administering at least one peptide as defined above or a nucleic acid encoding said peptide, or a composition comprising said peptide or nucleic acid, to an individual in need thereof, as described herein.
Also provided is a method of inducing a mixed mucosal IgG and IgA response to HIV-1, the method comprising administering at least one peptide as defined above or nucleic acids encoding said peptides, or a composition comprising said peptide or nucleic acid to an individual in need thereof, as described herein. Polypeptides include amino acid sequences modified either by natural processes, such as post-translational processing, or by chemical modification techniques which are well known in the art. Such modifications are well described in basic texts and in more detailed monographs, as well as in a voluminous research literature. Modifications can occur anywhere in a polypeptide, including the peptide backbone, the amino acid side- chains and the amino or carboxyl termini, it will be appreciated that the same type of modification may be present in the same or varying degrees at several sites in a given polypeptide. Also, a given polypeptide may contain many types of modifications. Polypeptides may be branched as a result of ubiquitination, and they may be cyclic, with or without branching. Cyclic, branched and branched cyclic polypeptides may result from natural posttxans!ational processes or may be made by synthetic methods. Modifications include acetylation, acylation, ADP-ribosylation, araidation, covalent attachment of flavin, covalent attachment of a heme moiety, covalent attachment of a nucleotide or nucleotide derivative, covalent attachment of a lipid or lipid derivative, covalent attachment of phosphatidyl inositol, cross-linking, cyclization, disulfide bond formation, demethylation, formation of covalent cross-links, formation of cystine, formation of pyroglutamate, formylation, gamma-carboxy!ation, glycosylation, GPI anchor formation, hydroxy! ation, iodination, mefhylation, myristoyl ation, oxidation, proteolytic processing, phosphorylation, prenylation, racemizaiion, selenoyi ation, sulfation, transfer-RNA mediated addition of amino acids to proteins such as arginyiation, and ubiquitination.
Variants of the peptide disclosed herein comprise peptides sequences of SEQ åDN°1 wherein 1 to 2 amino acids are substituted with other amino acids resulting in conservative substitutions. Exemplary conservative substitutions are listed below.
Ala (A) Val (V); Leu (L), lie (I)
Asn (N) Gin (Q); His (H); Lys (K)
Asp (D) Glu (E) lie (I) Leu (L); Val (V); Met (M); Ala (A)
Leu (L) Norleucine; He (I); Met (M); Ala (A); Phe (F)
Phe (F) Leu (L); Val (V); He (I); Ala (A); Tyr (Y)
Ser (S) Thr (T)
Trp (W) Tyr (Y); Phe (F)
The peptide vaccines of the invention may be formulated into a pharmaceutical form, preferably in combination with a pharmaceutically acceptable carrier. The pharmaceutical forms suitable for injectable use include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions in all cases the form must be sterile and must be fluid to the extent that easy syringability exists. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms, such as bacteria and fungi. The carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (e.g., glycerol, propylene glycol, and liquid polyethylene glycol, and the like), suitable mixtures thereof, and/or vegetable oils. Proper fluidity may be maintained, for example, by the use of a coating, such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants. The prevention of the action of microorganisms can be brought about by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like. In many cases, it will be preferable to include isotonic agents, for example, sugars or sodium chloride. Prolonged absorption of the injectable compositions can be brought about by the use in the compositions of agents delaying absorption, for example, aluminum monostearate and gelatin.
For parenteral administration in an aqueous solution, for example, the solution should be suitably buffered if necessary and the liquid diluent first rendered isotonic with sufficient saline or glucose. These particular aqueous solutions are especially suitable for intravenous, intraarterial, intramuscular, subcutaneous, intratumoral and intrap eritoneal administration. In this connection, sterile aqueous media that can be employed will be known to those of skill in the art in light of the present disclosure. For example, one dosage may be dissolved in I ml of isotonic NaCl solution and either added to 1000 ml of hypodermoclysis fluid or injected at the proposed site of infusion, (see for example, (see for example, Remington: The Science and Practice of Pharmacy, 21st Edition, Lippincot and Williams, 2005). Some variation in dosage will necessarily occur depending on the condition of the subject being treated. The person responsible for administration will, in any event, determine the appropriate dose for the individual subject. Moreover, for human administration, preparations should meet sterility, pyrogenicity, general safety and purity standards as required by FDA Office of Biologies standards.
Sterile injectable solutions are prepared by incorporating the active compounds in the required amount in the appropriate solvent with various of the other ingredients enumerated above, as required, followed by filtered sterilization. Generally, dispersions are prepared by incorporating the various sterilized active ingredients into a sterile vehicle which contains the basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile powders for the preparation of sterile injectable solutions, the preferred methods of preparation are vacuum-drying and freeze-drying techniques which yield a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.
The compositions disclosed herein may be formulated in a neutral or salt form. Pharmaceutically-acceptable salts, include the acid addition salts and which are formed with inorganic acids such as, for example, hydrochloric or phosphoric acids, or such organic acids as acetic, oxalic, tartaric, mandelic, and the like. Salts formed with the free carboxyl groups can also be derived from inorganic bases such as, for example, sodium, potassium, ammonium, calcium, or ferric hydroxides, and such organic bases as isopropylamine, trimethylamine, histidine, procaine and the like. Upon formulation, solutions will be administered in a manner compatible with the dosage formulation and in such amount as is therapeutically effective. The formulations are easily administered in a variety of dosage forms such as injectable solutions, drug release capsules and the like.
As used herein, ‘carrier’ includes, without limitation, solvents, dispersion media, vehicles, coatings, diluents, antibacterial and antifungal agents, isotonic and absorption delaying agents, buffers, carrier solutions, suspensions, colloids, liposomes and virosomes such as those described in Bomsel et al (2011) Immunity 34: 269-280. The use of such media and agents for pharmaceutical active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the active ingredient, its use in the therapeutic compositions is contemplated. Supplementary active ingredients can also be incorporated into the compositions.
The phrase ‘pharmaceutically-acceptable’ or ‘pharmacologically-acceptable’ refers to molecular entities and compositions that do not produce an allergic or similar untoward reaction when administered to a human. The preparation of an aqueous composition that contains a protein as an active ingredient is well understood in the art. Typically, such compositions are prepared as injectables, either as liquid solutions or suspensions; solid forms suitable for solution in, or suspension in, liquid prior to injection can also be prepared.
The vaccine composition will preferably contain an agent that enhances the protective efficacy of the vaccine, such as an adjuvant. Adjuvants include any compound or compounds that act to increase a protective immune response to the peptide antigen, thereby reducing the quantity of antigen necessary in the vaccine, and/or the frequency of administration necessary to generate a protective immune response. Adjuvants can include for example, emulsifiers; muramyl dipeptides; avridine; aqueous adjuvants such as aluminum hydroxide; oxygen-containing metal salts; chitosan-based adjuvants, and any of the various saponins, oils, and other substances known in the art, such as Ampfaigen, LPS, bacterial cell wall extracts, bacterial DNA, CpG sequences, synthetic oligonucleotides and combinations thereof (Schijns et al (2000) Curr. Opin. Immunol, 12:456), Mycohacterialplilei ( phlei) cell wall extract ( CWE) (U.S. Patent No. 4,744,984), M. phlei DNA (M-D A), and M-DNA-M phlei ceil wall complex (MCC), heat-labile enterotoxin (LT), cholera toxin (CT), cholera toxin B subunit (CTB). Compounds which can serve as emulsifiers include natural and synthetic emulsifying agents, as well as anionic, cationic and nonionic compounds. Oxygen-containing metal salts include salts of Al, K, Ca, Mg, Zn, Ba, Na, Li, B, Be, Fe, Si, Co, Cu, Ni, Ag, Au, and Cr which are sulphates, hydroxides, phosphates, nitrates, iodates, bromates, carbonates, hydrates, acetates, citrates, oxalates, and tartrates, and mixed forms thereof, including aluminium hydroxide, aluminium phosphate, aluminium sulphate, potassium aluminium sulphate, calcium phosphate, Maalox (mixture of aluminium hydroxide and magnesium hydroxide), beryllium hydroxide, zinc hydroxide, zinc carbonate, zinc chloride, and barium sulphateAmong the synthetic compounds, anionic emulsifying agents include, for example, the potassium, sodium and ammonium sails of lauric and oleic acid, the calcium., magnesium and aluminum salts of fatty acids, and organic sulfonates such as sodium lauryl sulfate. Synthetic cationic agents include, for example, cetyltrhethylammonlum bromide, while synthetic nonionic agents are exemplified by glycerylesters (e.g., glyceryl monostearate), polyoxyethylene glycol esters and ethers, and the sorbitan fatty acid esters (e.g., sorbitan monopalmitate) and their polyoxyethylene derivatives (e.g., polyoxyethylene sorbitan. monopalmitate). Natural emulsifying agents include acacia, gelatin, lecithin and cholesterol.
Other suitable adjuvants can be formed with an oil component, such as a single oil, a mixture of oils, a water-in-oil emulsion, or an oil-in- water emulsion. The oil can be a mineral oil, a vegetable oil, or an animal oil. .Mineral oils are liquid hydrocarbons obtained from petrolatum via a distillation technique, and are also referred to in the art as liquid paraffin, liquid petrolatum,, or white mineral oil. Suitable animal oils include, for example, cod liver oil, halibut oil, menhaden oil, orange roughy oil and shark liver oil, ail of which are available commercially. Suitable vegetable oils, include, for example, canola oii, almond oil, cottonseed oil, com oil, olive oil, peanut oil, safflower oii, sesame oil, soybean oil, and the like. Freund's Complete Adjuvant (FCA) and Freund's incomplete Adjuvant (FI A) are two common adjuvants that are commonly used in vaccme preparations, and are also suitable for use in the present invention. Both FCA and FIA are water-in-mineral oil emulsions; however, FCA also contains a killed Mycobacterium sp. Particularlay preferred adjuvants for mucosal vaccines include galactosyl ceramide (GalCer), as described in Lee et al (2011) Vaccine 29: 417-425.
Immunomodulatory cytokines can also be used in the vaccine compositions to enhance vaccine efficacy, for examplean adjuvant, Non-limiting examples of such cytokines include interferon alpha (IFN-a), interleukin-2 (IL-2), and granulocyte rnacrophage-colony stimulating factor (GM— CSF), or combinations thereof. GM-CSF is preferred.
When provided prophylactically, the immunogenic compositions of the invention are ideally administered to a subject in advance of HIV infection, or evidence of HIV infection, or in advance of any symptom due to AIDS, especially in high-risk subjects. The prophylactic administration of the immunogenic compositions can serve to provide protective immunity of a subject against HIV-1 infection or to prevent or attenuate the progression of AIDS in a subject already infected with HIV-1. When provided therapeutically, the immunogenic compositions can serve to ameliorate and treat AIDS symptoms and are advantageously used as soon after infection as possible, preferably before appearance of any symptoms of AIDS but may also be used at (or after) the onset of the disease symptoms.
Administration may be via a parenteral or non-parenteral route. Routes of administration will vary, naturally, with the location and nature of the disease, and include, e.g. intravenous, intrarterial, intradermal, transdermal, intramuscular, mucosal subcutaneous, percutaneous, intratracheal, intraperitoneal, perfusion and lavage. Preferably, administration is via a mucosal route, for example via a nasal, oral (via the mucosa of the digestive system), vaginal, buccal, rectal, sublingual, ocular, urinal, pulmonal or otolar (vie the ear) route.
For nasal administration, an exemplary formulation may be a nasal spray, lavage, drop or squirt system such as the Bidose Liquid from Aptar, Pfeffer Group or the Accuspray from Becton Dickinson (see Brandztaeg, 2011, AJRCCM, Bitter et al (2011) CurrProbl Dermatol 40: 20-35).
The treatments may include various ‘unit doses.’ A unit dose is defined as containing a predetermined-quantity of the therapeutic composition comprising a lentiviral vector of the present invention. The quantity to be administered, and the particular route and formulation, are within the skill of those in the clinical arts. A unit dose need not be administered as a single injection but may comprise continuous infusion over a set period of time. A unit dose may contain at least 0.01, 0.05, 0.1, 0.5, 1.0, 2.5, 5.0, 10.0, 15.0, 25.0 or 50.0 mg of the active ingredient. Optionally, a unit dose contains less than 0.01, 0.05, 0.1, 0.5, 1.0, 2.5, 5.0, 10.0, 15.0, 25.0 or 50.0 mg of the active ingredient. In one embodiment, a unit dose contains from about 0.001 mg to about 50 mg of the active ingredient. In another embodiment a unit dose contains from about 1 mg to about 10 mg of active ingredient.
In one embodiment, the vaccine composition may be administered in a single daily dose, or the total daily dosage may be administered in divided doses, for example, two, three or four times daily. Furthermore, the vaccine composition may be administered in intranasal form via topical use of suitable intranasal vehicles, via transdermal routes, using those forms of transdermal skin patches well known to persons skilled in the art, by implantable pumps; or by any other suitable means of administration. To be administered in the form of a transdermal delivery system, for example, the dosage administration will, of course, be continuous rather than intermittent throughout the dosage regimen.
The dosage regimen utilizing the vaccine composition is selected in accordance with a variety of factors including type, species, age, weight, sex and medical condition of the patient; the severity of the condition to be treated; the route of administration; the renal and hepatic function of the patient; and the particular compound employed. A physician or veterinarian of ordinary skill can readily determine and prescribe the effective amount of the drug required to prevent, counter or arrest the progress of the disease or disorder that is being treated.
Vaccine administration may further comprise a prime-boost regimen. In these methods, one or more priming immunisations are followed by one or more boosting immunisations. The actual immunogenic composition can be the same or different for each immunisation and the type of immunogenic composition, the route, and formulation of the immunogens can also be varied. One useful prime-boost regimen provides for two priming immunisations, four weeks apart, followed by two boosting immunisations at 4 and 8 weeks after the last priming immunisation.
Such a regimen may comprise, for example, priming with full-length gp41 and boosting with one or more immunogenic peptides as disclosed herein. Either the prime or the bosst, or both, may be administered in the form of a DNA molecule encoding the peptide or polypeptide in question. Without being bound by theory, it is thought that this regimen might elicit gp41 cross-reactive antibodies targeted to the epitopes present in the priming immunogen. The designed protein fragments may be expressed in E. coli in order to prevent glycosylation and consequent epitope masking that might occur if expressed in a eukaryotic expression system. The use of E. coli to produce non-glycosylated versions of the invention may have contributed to the success of the approach.
Immunisation schedules (or regimens) are well known for animals (including humans) and can be readily determined for the particular subject and immunogenic composition. Hence, the immunogens can be administered one or more times to the subject. Preferably, there is a set time interval between separate administrations of the immunogenic composition. While this interval varies for every subject, typically it ranges from 10 days to several weeks, and is often 2, 4, 6 or 8 weeks. For humans, the interval is typically from 2 to 6 weeks. In a particularly advantageous embodiment of the present invention, the interval is longer, advantageously about 10 weeks, 12 weeks, 14 weeks, 16 weeks, 18 weeks, 20 weeks, 22 weeks, 24 weeks, 26 weeks, 28 weeks, 30 weeks, 32 weeks, 34 weeks, 36 weeks, 38 weeks, 40 weeks, 42 weeks, 44 weeks, 46 weeks, 48 weeks, 50 weeks, 52 weeks, 54 weeks, 56 weeks, 58 weeks, 60 weeks, 62 weeks, 64 weeks, 66 weeks, 68 weeks or 70 weeks.
The immunisation regimes typically have from 1 to 6 administrations of the immunogenic composition, but may have as few as 1, 2, 3, 4 or 5. The methods of inducing an immune response can also include administration of an adjuvant with the immunogens. In some instances, annual, biannual or other long interval (5-10 years) booster immunisation can supplement the initial immunisation protocol.
A specific embodiment of the invention provides methods of inducing an immune response against HIV in a subject by administering an immunogenic composition of the invention, , one or more times to a subject wherein the peptic sequence is expressed at a level sufficient to induce a specific immune response in the subject. Such immunisations can be repeated multiple times at time intervals of at least 2, 4 or 6 weeks (or more) in accordance with a desired immunisation regime.
The immunogenic compositions of the invention can be administered alone, or can be co-administered, or sequentially administered, with other HIV immunogens and/or HIV immunogenic compositions, e.g., with ‘other’ immunological, antigenic or vaccine or therapeutic compositions thereby providing multivalent or ‘cocktail’ or combination compositions of the invention and methods of employing them. Again, the ingredients and manner (sequential or co-administration) of administration, as well as dosages can be determined taking into consideration such factors as the age, sex, weight, species and condition of the particular subject, and the route of administration.
When used in combination, the other HIV immunogens can be administered at the same time or at different times as part of an overall immunisation regime, e.g., as part of a prime-boost regimen or other immunisation protocol.
Many other HIV immunogens are known in the art. One such immunogen is HIVA (described in WO 01/47955), which can be administered as a protein, on a plasmid (e.g., pTHr.HIVA) or in a viral vector (e.g., MVA.HIVA). Another such HIV immunogen is RENTA (described in PCT/US2004/037699), which can also be administered as a protein, on a plasmid (e.g., pTHr.RENTA) or in a viral vector (e.g., MVA.RENTA).
For example, one method of inducing an immune response against HIV in a human subject comprises administering at least one priming dose of an HIV immunogen and at least one boosting dose of an HIV immunogen, wherein the immunogen in each dose can be the same or different, provided that at least one of the immunogens is an epitope of the present invention, a nucleic acid encoding an epitope of the invention or an expression vector, preferably a VSV vector, encoding an epitope of the invention, and wherein the immunogens are administered in an amount or expressed at a level sufficient to induce an HIV-specific immune response in the subject. The HIV-specific immune response can include an HIV-specific T-cell immune response or an HIV-specific B-cell immune response. Such immunisations can be done at intervals, preferably of at least 2-6 or more weeks.
‘Treatment’ includes both therapeutic treatment and prophylactic or preventative treatment, wherein the object is to prevent or slow down the targeted pathologic condition or disorder. Those in need of treatment include those already with the disorder as well as those prone to have the disorder or those in whom the disorder is to be prevented. The terms ‘therapy’, ’therapeutic’, ‘treatment’ or ‘treating’ include reducing, alleviating or inhibiting or eliminating the symptoms or progress of a disease, as well as treatment intended to reduce, alleviate, inhibit or eliminate said symptoms or progress. Desirable effects of treatment include preventing occurrence or recurrence of disease, alleviation of symptoms, diminishment of any direct or indirect pathological consequences of the disease, preventing metastasis, decreasing the rate of disease progression, amelioration or palliation of the disease state, and remission or improved prognosis. In some embodiments, methods and compositions of the invention are used to delay development of a disease or disorder or to slow the progression of a disease or disorder.
Preferably, an effective amount, preferably a therapeutically effective amount of the (poly)peptide or vector of the invention is administered. An ‘effective amount’ refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired therapeutic or prophylactic result. The effective amount may vary according to the drug or prodrug with which the (poly) peptide or vector is co-administered.
A ‘therapeutically effective amount’ of a (poly) peptide or vector of the invention may vary according to factors such as the disease state, age, sex, and weight of the individual, and the ability of the (poly) peptide, to elicit a desired therapeutic result. A therapeutically effective amount encompasses an amount in which any toxic or detrimental effects of the (poly) peptide are outweighed by the therapeutically beneficial effects. A therapeutically effective amount also encompasses an amount sufficient to confer benefit, e.g., clinical benefit.
Throughout the specification, terms such as ‘comprises’, ‘comprised’, ‘comprising’ and can have the meaning attributed to them in most patent jurisdictions, preferably in the jurisdiction in question; e.g. they can mean ‘includes’, ‘included’, ‘including’, etc. Terms such as ‘consisting of ‘consisting essentially of and ‘consists essentially of have the meaning ascribed to them in most patent jurisdictions, preferably in the jurisdiction in question; e.g., they may imply the exclusion of all, most, or all but a negligible amount of other elements, or they may allow for elements not explicitly recited, but exclude elements that are found in the prior art or that affect a basic or novel characteristic of the invention.
An ‘isolated’ peptide or nucleic acid may be isolated substantially or completely away from one or more elements with which is associated in nature, such as other naturally occurring peptide or nucleic acids or other peptide or nucleic acid sequences. The invention will now be described in more detail by means of the following non limiting figures and examples. All patent and literature references cited herein are hereby incorporated by reference in their entirety.
FIGURES:
FIGURE 1. FabA 43 conformational epitope P7 designed in silico blocks FabA 43 binding to gp41 and reverse FabA 43 neutralization activity. A : Preincubation of FabA 43 with the conformational epitope P7 (5 mM, hatched bars) but not with control HA (5mM, plain bars) interferes significantly with FabA 43 binding to clade A, clade B and clade C PI or clades A and C gpl40 and clade B gp41, as evaluated by ELISA. B : The P7-induced reduction of FabA 43 binding to each antigen is dose dependent. Binding inhibition to FabA 43 binding to each antigen in the presence of P7 is calculated relative to the binding inhibition in the presence of the irrelevant HA peptide serving as negative control. In A and B, values represent mean ± SEM, derived from 3 independent experiments performed in triplicate. C : Localization of the amino acids paths corresponding to P7 peptide on the 6-Helix bundle conformation of clade A, B and C gp41 framel5. Each gp41 monomer of the trimer is depicted using a different tone of gray. The amino acid paths corresponding to the FabA43 specific peptide P7 are highlighted in orange. Note residues 532-535 belong to the N-Helix of one monomer while residues 669- 672 belong to the C-Helix of another monomer of the trimer. Red: the P7 predicted conformation superimposed on the structure of gp41. RMSD values over the paired residues of gp41 and P7 are of 2.45, 2.97 and 1.97 A for clade A, B and C, respectively. Note that in this region of the gp41, only residue 535 differs between clade B in one hand and clade A and C in the other hand. D : Conformational variability of the predicted conformations of P7 using PEP-FOLD3. Left: conformation best fitting gp41. Right: top 10 predicted conformations superimposed. E : Interference of FabA 43 neutralization by a 400-fold molar excess of P7 peptide. The percentage of neutralization, analyzed by flow cytometry as indicated in Materials and methods section, is shown relative to FabA 43 neutralization using primary CD4+ T cells in the presence of the irrelevant HA peptide, tested in parallel. Values represent mean ± SEM, derived from 3 independent experiments performed.
FIGURE 2: P7 neutralisation of HIV infection of CD4+T cells
P7 induce inhibition of CD4+T cells infection in a dose-dependent manner. The potential of P7 to inhibit CD4+T cells infection was assessed in a single cycle infectivity assay, using p24 staining on primary CD4+T cells. P7, at indicated concentrations, was preincubated for 1 h at 37° with either free virus (dotted line, diamond) or activated CD4+T cells (dotted line, triangle). After lh, activated CD4+T cells were added when P7 was preincubated with the virus, or virus was added when P7 was preincubated with CD4+T cells. Cultures were then incubated for 36 h. The percentage of neutralization, analyzed by flow cytometry, was defined as the reduction of the Gag-p24+ CD4+T cells in the presence of P7 compared with HIV- 1 -infected CD4+T cells in the absence of P7 (solid line, circle). Values represent mean ± SEM, derived from at least 4 independent experiments performed in triplicate.
FIGURE 3: P7 binds to trimeric envelope gpl40 cross clade
A. Binding of P7 (at increasing concentration up to 20 mM) to HIV-1 envelope was evaluated by ELISA after immobilization of gpl40 of either clade A (plain black bars), B (white bars) and C (hatched bars). B. The irrelevant 9 amino acid influenza virus Hemagglutinin (HA) peptide (YPYDVPDY) at 20 mM was used as a negative control. Peptide binding was detected with FabA 43 followed by HRP coupled anti-human IgA. n= 3 independent experiments. Student’s t-test, * = p<0.05 ***; = p<0.001
MATERIALS AND METHODS
- Proteins and Peptides
The gp41 (584-684) used was a construct based on the HXB2 group M subtype B sequence [40] and kindly provided by Dr. Weissenhorn. Gpl40 from HIV-1 clades A (92RW020) and C (C.ZA.1197MB) were obtained from Immune Technology Corp (NY10021, USA). Sequences of PI (a.a 630-685) derived from clade B HXB2 gp41 (Pl-B) was SQTQQEKNEQELEELDKWASLWNWFDITNWLWYIK (SEQ IDN°2) as described [22], from clade A 99UGA07072 gp41 (PI -A) was SQIQQKKNEQDLLALDKWANLWNWFDISNWLWYIR (SEQ IDN°3) and from Clade C Bw96Bw0502 (Pl-C) was SQTQQEKNEQELLALDS WKNLWNWF SITNWLWYIK (SEQ IDN°4). Peptides were chemically synthesized at a purity >95% by Biopeptide (LA, USA) for Pl-B and United BioSystems (VA, USA) for Pl-A and -C.
- Cloning of FabG and Production and Purification of soluble FabA and G
ESN Fab As were transformed in their corresponding FabGs by molecular cloning using pASK88 vector which direct the synthesis of the human gamma- 1 heavy chain and light chain, respectively [41] Production of functional Fabs was performed as previously described [2] Briefly, cultures were grown in 1L of LB medium containing 100pg/ml ampicillin and expression was induced for 14 hr at 22°C by addition of 0.2 mg/L of anhydrotetracycline (ACROS Chimica) at an OD550 of 0.5. Fabs were purified from the periplasmic fraction of the E. coli pellet by immobilized metal affinity chromatography (IMAC) using a GE Healthcare kit (GE Healthcare, Sweden, 17-5286-01, HisTrap FF crude). After elution from the affinity media with 0.5M imidazole, the FabA and G were dialyzed against PBS and concentrated using Amicon Ultra- 10 centrifugal filter units (Millipore). Antibody purity was evaluated by native SDS/PAGE, followed by Coomassie blue staining resulting in a single band at 50 kDa corresponding to the Fab heterodimer comprising the heavy and light chains, as expected. No band at 25 kDa corresponding to either isolated Fab light or heavy chain could be detected.
The concentration of FabA and G was measured by sandwich ELISA allowing the restricted detection of full Fabs, namely covalently linked heterodimers, using goat anti human IgA or IgG (Caltag, France) for coating, and biotinylated mouse anti-human Ig kappa light chains (B.D. Pharmingen, USA) for detection, as earlier described [2]
- Binding assays
Surface Plasmon Resonance (SPR): All experiments were performed in duplicate, as described in [11, 42] with a Biacore 3000 instrument (Biacore, Inc.) at 20°C in HBS-EP running buffer [10 mM Hepes (pH 7.4), 150 mM NaCl, 3 mM EDTA, 0.005% surfactant P20] Immobilization of various HIV-1 envelope subunit ligans ligands, namely Clade B gp 41, Clade A and C gpl40, and clade A, B and C PI peptides, to CM5 chips (GE Healthcare) followed the standard procedures recommended by manufacturer. The final immobilization levels were between 300 and 500 RU to avoid rebinding events, as mentioned in [42] Initial binding experiments with clone 43 and 177 of FabA and G, and non-specific isotypes as analytes were comparatively performed. No specific signal was observed with non-specific isotypes (recorded value of 0 RU), indicating that the observed binding of Fabs the different HIV-1 envelope subunits was specific. For kinetic measurements, sensorgrams were obtained by passing various concentrations of the analytes at various concentrations as indicated, over the ligand surface, at a flow rate of 50pl/min, with a 3-min association phase and a 6-min dissociation phase. The sensor surface was regenerated between each experiment with a single injection of 35 mM NaOH and 1.3 M NaCl at a flow rate of 50 mΐ/min for 30sec. Identical injections over blank surfaces ran in parallel (and giving a value of 0 RU) were subtracted from the data for kinetic analysis. Binding kinetics were evaluated by linearization using “fit kinetics Langmuir binding type” with BiaEvaluation software (BiacoreTM). Relative Pearson's chi- squared tests assessing goodness of fit were always below 10, which indicated that the models used for fitting adequately describe recorded data.
ELISA: ELISA binding assays were performed as described [11] by coating microtiter plates (NUNC-Immuno Plate MaxiSorp Surface, or Peptide Immobilizer Exiqon Peptide Immobilizer, Exiqon) with gpl40 (trimeric rgp41 at 0.25 microg/well), PI (0.1 pg/well), peptides corresponding to conformational epitopes (0.1 pg/well), overnight at 4°C in PBS. Fab binding was detected with a biotinylated mouse anti-human Ig kappa light chains (BD Pharmingen) and streptavidin- HRP or with anti-human IgA HRP (Jackson ImmunoRe search) for the peptides corresponding to conformational epitopes. All experiments were performed with Fabs from at least three independent purifications, each in duplicate. For competition ELISA, Fab 43 was preincubated ON at 4°C with various concentration of P7 or the irrelevant Hemagglutinin (HA) (YPYDVPDY) peptide serving as negative control and added to gp41 clade B, gpl40 (clades A, C) or PI (clades A, B, C) coated on the ELISA plate at a final concentration of 0.8nM for FabA 43 and 0.05 to 1 OmM for P7. Fab binding to each gp41 subunit was finally detected enzymatically by using anti human IgA coupled with HRP.
Fab binding to HIV-1 infected cells: Binding assays were performed as described [43] For detection of Fab binding to native HIV-1 envelope at the surface of HIV-1 infected cells, FabA or G (5pg/ml) were incubated with 105 cells overnight at 4°C. To allow for direct isotype comparison, FabA and G were detected in parallel using the same fluorescein isothiocyanate (FITC)-conjugated mouse anti-human kappa light chain (BD Biosciences, San Jose CA, USA), 30 minutes at 4°C. Cells were next fixed with 4% paraformaldehyde, and were further stained for intracellular Gag-p24 with the phycoerythrin (PE) coupled anti-Gag KC57 murine monoclonal antibody (Beckman Coulter GmbH). Fab binding to target cells was quantified by flow cytometry using a Guava EasyCyte flow cytometer (Merck-Milipore), and analyzed using the dedicated InCyte software.
- Target cells
Peripheral blood samples from healthy donors, obtained from the Etablissement Franqais de Sang (Paris, France) were depleted of CD8+T cells with Rosette Sep cocktail, (StemCell Technologies Inc., France) and peripheral blood mononuclear cells (PBMC) were isolated by Ficoll-Hypaque. After stimulation for 2 days with 5pg/ml phytohemagglutinin (Sigma-Aldrich, St.Louis, MO) as described [11], CD8-depleted PBMCs were used for infection and neutralization experiments. Alternatively, CD4+T CEM-NKR lymphocytic cells (NK-resistant) expressing CCR5 (AIDS Research and Reference Program, NIH) we used as when indicated. To prepare CD4+ T target cells for neutralization assays, cells were split 1 :3 on the day of passage and used the following day.
Primary monocytes were purified from peripheral blood samples from healthy donors using human monocyte enrichment kits (StemCell Technologies Inc., France) and differentiate into Langerhans cell with TGF-b, GM-CSF and IL-4, as described [11]
- Virus stock preparation
A stock of HIV-1JR-CSF (clade B, R5 tropic) was prepared on a large scale by transfecting 293T cells with a plasmid containing the DNA sequence of JR-CSF (NIH, Germantown, MD USA) [2] The cell culture supernatant was concentrated, separated into single use aliquots and stored at -80 deg C.
The HIV-1 primary isolates 92UG031 (clade A, R5) and 93BR025 (clade C, R5), obtained through the NIH AIDS Reagent Program, was amplified on PBMCs, as previously described [2] Virus concentration was quantified by measuring p24 antigen by ELISA (Innotest HIV-1 Antigen mAh, Innogenetics).
- HIV-1 Neutralization Assays
Single-cycle neutralization assay: The neutralization activity of FabA and FabG was evaluated on primary CD4+ T-cells (CD8+T cells-depleted PBMC), on CEM-CCR5+ infected with HIV-1 JR-CSF (Clade B) or with each of Clade A or C primary isolates and quantified by flow cytometry after intracellular Gag-p24 staining, as we described earlier [2] At least five independent experiments, performed each in triplicate, were performed. Live cells initially gated by forward and side scatter were analyzed for intracellular expression of p24-Ag. A dose dependent parameter was used to compare the FabA with FabG and for determination of maximum percent inhibition values. Neutralization was defined in % of cells infected in the absence of antibody. Titers were calculated as IC50 and IC85 and reported as the concentration of Fab causing a 50 or 80% reduction in the percentage of p24+ cells compared to virus control wells.
For competing the neutralization, FabA 43 was preincubated with a 400-fold molar excess of P7 overnight at 4°C and further incubated the virus and the target primary CD4+ T-cells at final FabA 43 concentration of 20nM and P7 concentration of 33 mM. The irrelevant peptide from the Hemagglutinin HA (YPYDVPDY) serving as negative control had no effect on FabA 43 neutralizing activities.
Inhibition of HIV-1 transfers from cells to cells. The inhibition of HIV-1 transfer from Langerhans cells to T cells was evaluated using monocytes derived Langerhans cells (LCs) obtained from PBMCs, and autologous TCD4+ cells as we previously described [11] Briefly, LCs were incubated with either clade of HIV-1 for 2hrs at 37°C, washed extensively to remove the free virus and distributed in 96 well plates at 100,000 cells / well. Indicated concentrations of FabA or G were then added to the corresponding wells. Finally, either medium alone, or resting TCD4+ cells in medium (100,000 cells / well) were added. The LCs-T co-cultures were incubated at 37°C for 5 days. The virus transfer was evaluated by measuring Gag-p24 released in the culture medium using a commercial ELISA (Innotest HIV-1 Antigen mAb, Innogenetics) according to manufacturer instructions. Results are expressed as % of inhibition transfer using formula [(LC+T-Ab) - (LC+T+Ab) / (LC+T-Ab)] xlOO.
- Epitope mapping
Epitope mapping of both isotypes was performed as we described previously [11], using linear 12-mer peptide libraries displayed on the protein pill of M13 phages (New England Biolabs) as recommended by the manufacturer. In brief, IgA- or IgG-coated beads were incubated with Fab IgA or IgG on a rotating wheel for 2 h at room temperature and epitope screening was initiated by incubating each bead of Fab IgA or IgG with the original 12-mer (1013) phage displayed peptide library containing different phages, overnight at 4°C. After three rounds of selection with increasing stringency, the phages were tittered, and single clones were picked and tested by phage ELISA for specific binding on each FabA or FabG. Positive clones were amplified, and each specific peptide insert was sequenced. Importantly, two round of negative selection using beads coated with normal human IgA or IgG (Jackson ImmunoRe search) were introduced between two rounds of positive selections. Phages remaining from the negative selection were amplified in Escherichia coli ER2738, precipitated, and used for second and third rounds of selection similar to the first round, but with increased buffer stringency as described [11]
- Modeling clade A, B and C gp41
Six Helix bundle early post fusion clade B gp41: X-ray crystallography studies have been highly successful in giving information about the helical regions of the HIV-1 gp41 ectodomain in its 6-Helix bundle state; however, in all cases to date the loop region was either removed to facilitate crystallization or not visible in the crystal structure [44]; [33]). However, the structure of the SIV gp41 ectodomain containing the loop region was successfully determined by NMR spectroscopy [45] The HIV-1 and SIV loops share a high degree of sequence identity (46%). The structure of the trimeric gp41 ectodomain of HIV-1 in its 6-Helix bundle state was thus built using the X-ray crystallography data available for the six-helix bundle of HIV- 1, and the NMR data available for the loop of SIV using the MODELLER v9.15 software [46] The conserved disulfide bond of the loop between residues C68 (C599) and C74 (C605) was added as a constraint. This bond was proved to be critical to the furin recognition site of HIV- 1 gpl60 [47]
The crystal structure of the 6-Helix bundle HIV-1 gp41 including both fusion peptide and membrane proximal external regions [33] was retrieved from the RCSB Protein Data Bank (http://www.rcsb.org, code : 2X7R). The solution NMR structure of ectodomain of SIV gp41 [48] was retrieved from the RCSB Protein Data Bank (http://www.rcsb.org, code : 2EZO). Multiple sequence alignment was used to build the 6- Helix bundle structure of HIV- 1 gp41.
Pre fusion clade B gp41: The structure of the trimeric gp41 ectodomain of HIV- 1 in its pre-fusion state was built using the sole crystallographic structure of the pre-fusion HIV-1 gp41 and missing loops were modeled using the BCLoopSearch algorithm (http://bioserv.rpbs.univ-paris-diderot.fr/services/BCSearch/) which helps identify linear fragments similar to a query in large collections of structures using a new similarity approach based on a Binet Cauchy (BC) kernel [49]
The crystal structure of the pre-fusion HIV-1 gp41 [32] was retrieved from the RCSB Protein Data Bank (http://www.rcsb.org, code : 4TVP).
Pre and post fusion Clade A and C gp41: The clade A and clade C gp41 sequences were retrieved from http://www.hiv.lanl.gov (code : AF484478 and code : AF110967, respectively) aligned and mapped onto the previous structures using the MODELLER v9.15 software [46] We used all atom molecular dynamics (MD) simulations to inspect the behavior of the 6 trimeric structures (pre-fusion : clade A, B and C, 6-Helix bundle : clade A, B and C) and explore their conformational space. The simulations were performed under periodic boundary conditions by defining a box with a minimal distance of 1.0 angstroms between the protein and the wall of the cell. The box was then filled with water and we added a concentration of 0,2 M Na+Cl- to the solvent, as well as an excess of Na+ ions to neutralize the system charge induced by the negatively charged residues. 200 ns of Molecular Dynamics simulations were then run for the structures using Gromacs 2016.1 [50] [51] with AMBER99SB-ILDN [52] as force field and TIP3P [53] model for water.
- Mimotope Sequence Analyses and docking on gp41
For each set of Fab A and FabG mimotopes, epitope mapping on pre and post fusion clade A, B and C gp41 was performed by computational method that aims to predict discontinuous or 3D eptitopes. Three available computational methods to map mimotopes sequences to conformational epitopes were evaluated: PepSurf, Mapitope [54] and [55] We selected PepSurf for this study because it provided the best results - as it was able to retrieve the PI epitope on both pre-fusion and 6-Helix bundle structures using mimotopes of 2F5 IgG and IgA - and allowed the user to define the similarity matrix. A discontinuous 3D epitope is localized on the protein surface by searching for a 3D fit with partial amino acid strings of a given sequence in a preset distance. PepSurf compares each peptide provided to the solvent accessible surface of the antigen, determining the best path. The algorithm then creates clusters of antigen residues on the surface that best fit a grouping of peptides and returns a score for each residue at the surface corresponding to the number of occurrences of this residue in the alignments.
Snapshots of the gp41 trimeric structures were taken every 10 ns of the MD starting from 60 ns and explored using the pepSurf algorithm with both BLOSUM62 and BLOSUM80 as similarity matrix and the 141 antibody binding mimotopes as input. A library of 147 12-mer soluble peptides was generated using SolyPep on the RPBS web portal and used also as input to account for the background noise. Scores were then averaged for each residue position for the three chains of the 15 structure snapshots to account for the protein flexibility. Scores obtained for the random peptides library were subtracted.
Using this method, we found three regions that could correspond to conformational 3D epitopes on the 3 6-Helix bundle structures:
• G1-Q10 and E/A132-F143 which correspond to the N-terminus of the first helix and the C-terminus of the second helix of each monomer, respectively,
• E30-T39 and 1105-1/Ll 16 which are roughly located in the middle of both helices,
• Q60-W66 and A/N77-A/S83 which encompass the top of the helices and the loops. Results for the 3 pre-fusion structures were less clear but highlighted a region in the vicinity of the PI epitope (A/E132-F143) and a region near the epitope found in the 6- Helix bundle structure at A/N77-A/S83.
Although PepSurf consider the relative distance and physicochemical properties of individual amino acids, it does not take into account the 3D conformation that the mimotopes might adopt in the solvent. The 141 tested mimotopes that encompass the found epitopes were thus modeled using the PEP-FOLD3 algorithm, a de novo approach aimed at predicting peptide structures from amino acid sequences, based on structural alphabet letters [56] The peptide structures were then aligned to the paths found by the PepSurf algorithm on all 6 structures of gp41 (pre-fusion : clade A, B and C, 6-Helix bundle : clade A, B and C) by a pair-fit procedure to segregate paths that comply with the 3D conformation. Conformational paths - that is, paths that encompass two different chains - were selected and the resulting sequence modeled with the PEP-FOLD3 algorithm. Only peptide structures that aligned to the same sequence in the gp41 structure with a RMSD lower than 2,5 angstroms were retained, in order to keep only the peptide that would fold in water in the same manner as in the gp41 structures.
- Statistical analysis
Statistical significance was analyzed by the two-tailed Student’s t-test and multiple comparisons were performed using the non-parametric Mann- Whitney U test using Prism 5 (GraphPad, San Diego, CA) software. Heatmap was established using the heatmaper web site (http://www.heatmapper.ca/expression/) using the complete linkage clustering method and non-parametric spearman correlations as distance measurement method. P values <0.05 were considered significant.
RESULTS
1.1. CHI Isotype switching of mucosal FabA into FabG
To evaluate the role of the CHI domain in mucosal Ab specificity and function, we took advantage of the mucosal anti-HIV-1 FabA derived from cervical B cells of ESN individuals we previously characterized [2] Two FabA clones, namely 43 and 177, were obtained after screening of the ESN library on the clade B gp41 membrane proximal extended region (MPER) PI [18] [19] or using a recombinant Clade B gp41 deleted for its MPER [2], respectively. At the molecular level, FabA 43 has a high degree of somatic mutations, a VH3 heavy chain origin similar to the broadly neutralizing IgG 4E10 [20], and a long CDRH3 of 22 residues, features characteristics of other broadly neutralizing IgGs [2] In contrast, FabA 177 has a low level of somatic mutations for their heavy chain, 100% homology with germ-line gene region IGKVID-30*-01, and a normal length CDRH3 of 11 residues [2] Furthermore, FabA 177 heavy chain origin is VH6 family, which was never described before for broadly neutralizing IgGs. Each FabA was transformed by genetic engineering replacing the CHlal into CHlyl , as described in the method section, in their corresponding FabG, the two isotypes thus sharing identical VH and VL domains and the same light chain, but expressing different CHI domain.
1.2. FabA and FabG binding and affinity for HIV-1 envelope
We first evaluated whether class- switching preserves the original binding properties of IgA by comparing FabA and FabG binding to recombinant trimeric gp41 derived from HIV-1 Clade B [19] and gpl40 derived from HIV-1 Clades A and C. To avoid bias introduced by different isotype specific detection antibody, binding to gp41 in ELISA was detected using mouse anti-human kappa light chain [11] FabA 43 and 177 recognize conformational conserved regions on clade B gp41 from both R5 and X4 tropic viruses [2] Here, we found that both FabA and G pairs specifically bind to Clade B gp41 as well as to Clade A and C gpl40 in a dose-dependent manner, but strikingly for all clades, binding of FabA 43 and 177 is more efficient compared to their respective FabG Hence, a 50 times higher concentration of FabG 43 (50pg/ml) is required to reach the same binding of FabA 43 (lpg/ml)). Differences observed between FabA and G 177 is even larger, reaching 500 between the isotypes in favor of FabA. Similarly, binding to clade A and C gpl40 requireslO times less FabA 43 than FabG 43 and 50-100 times less FabA 177 than FabG 177. Importantly, these results show that both ENS clones recognize the viral envelope cross clade.
Surface plasmon resonance (SPR) was next used in kinetic experiments to compare of FabA and G affinity to clade B gp41 or clade A or C gpl40 immobilized onto the sensor ship surface. FabA and G 43 and 177 at various concentrations were the analytes. As a result (Table 1), when Clade B gp41 serves as target, KD is 3.62 nM for FabA 43 and 21.2nM for FabA 177 compared to 1.12 and 1.22 mM for their respective FabG. When Clade A and C gpl40 serve as target, KDs are 6.41 and 4.79nM for the FabA 43 compared to 0.50mM and no affinity is measured for its respective FabG; KD equals 2.92 and 2.15nM for FabA 177 whereas its corresponding FabG has no measurable affinity for any of the clade A or C viral envelope. Overall, affinities reach the nM range for the FabA, whereas remain in the order of mM or even lower for the FabG for both clones, in agreement with binding experiments using ELISA and show that the affinity of both FabA 43 and 177 are greater, for all clades, than that of their FabG counterpart. Importantly, for all Fab taken together, affinity correlates with binding observed by ELISA (spearman r= 0.606; p=0.375)
Altogether, FabA 43 and 177 bind to clade B gp41 and clade A and C gpl40 with nM affinity whereas affinities of corresponding FabG are much lower, although still able to bind their targets as shown by ELISA approaches.
Figure imgf000026_0001
1.3. Binding of FabA and FabG to the PI peptide
The 35 amino acid peptide PI was characterized as the minimal membrane proximal external region of the viral envelope gp41 that allows for HIV-1 binding to galactosyl ceramide, the HIV-1 mucosal receptor and for mucosal HIV-1 entry by transcytosis [21] [22] [23] This highly conserved region has been used as immunogen in a prophylactic vaccine against HIV-1 both in pre-clinical and clinical phase I trials [19, 24] PI sequence is well conserved between clade B and A viruses whereas a K670S mutation in clade C virus prevents binding of the broadly neutralizing 2F5 IgG [25] FabA 43 was obtained by screening the FabA ESN library on clade B PI and consequently FabA 43 binds to clade B PI [2] We thus evaluated by ELISA the capacity of FabA and G 43 to bind to clade A, B and C PI, comparatively as described [2] Both Fab 43 isotypes bind to clade A, B and C PI in a dose-dependent manner. Strikinly, as above when the PI peptide is expressed in the context of a full protein, FabA 43 recognizes PI more efficiently than its corresponding FabG. This difference is higher when clade B PI is the target (with a 60- fold change from FabA to G), but still significant (with a 20-fold change from FabA to G) when Clade A and C are targets. Additionally, FabA and G 43 bind more efficiently to Clade B compared to Clade A and C PI. These differences in Fab isotype binding to PI from the three clades are also observed by SPR.
Overall, the FabA 43 and 177 affinities for the gp41 viral envelope cross clade is in the nM range, similar to that of other broadly neutralizing Abs (bNAbs) [26] whereas FabG recognize less efficiently the same antigen.
1.4. FabA and FabG binding to HIV-infected CD4+ T-cells
We next evaluated the capacity of Fabs to bind to the viral envelope in its trimeric spike conformation at the surface of CD4+ T-cells infected with clade A (primary isolate 92UG031), B (isolate JR-CSF) or C virus (primary isolate 92BR025). After overnight incubation at 4°C with the various Fabs, binding was evaluated by flow cytometry using an anti-kappa light chain secondary antibody to allow direct isotype comparison. All Fabs bind specifically HIV-1 infected cells although recognition of Clade C and clade A HIV-1 infected cells by FabG 43 and FabG 177 is low. Irrelevant IgA and IgG used as negative controls give negligible binding signal. As observed for recombinant viral protein binding, for all clades, FabA 43 and 177 bind HIV-1 infected cells better than their respective FabG with 20-40% of T-cells labelled for FabA compared to 5-19% for FabG (Student’s t -test, p<0.05) for Fab 43 and 177 respectively). FabA 43 recognizes more efficiently Clade A than clade B and C infected CD4+ T-cells (Student’s t -test, p<0.05), whereas no differences were apparent between clades for FabA 177. Importantly, Fab binding to HIV- 1 infected cells correlates with Fab affinity and binding to HIV-1 envelope measured by ELISA (spearman r= 0.777 p=0.0156).
1.5. FabA and FabG antiviral properties
As FabA and G only differ by the CHI domain, the results presented above suggest that the CHI domain can affect the paratope and its fitting on the antigen and may in turn impact the Ab anti-viral function in an isotype-dependent manner.
- Neutralization of HIV-1 CD4+ T-cell infection by FabA and FabG
We first investigated the participation of the CHI domain in Fab isotype neutralizing activities, comparatively. Using a panel of three viruses from Clade A, B and C, neutralization of infection by FabA and G pairs of both 43 and 177 clones of CD4+T lymphocyte was tested, as previously described [2] Serial dilutions of each FabA and G were assessed comparatively, and IC50 values were determined for each HIV-1 clade. Comparative evaluation of paired Fab 43 isotypes reveals that FabA43 neutralizes all three A, B and C clades with respective IC50 of lng, lpg and lOOng/ml, whereas FabG 43 only neutralizes clade A virus with an IC50 of lOOng/ml and not Clade B or C virus clades. A similar neutralization pattern is observed for the clone 177 with an IC50 of lng, 300ng and 30ng/ml for FabA 177 against Clade A, B and C virus respectively and an IC50 of lOng and lpg for FabG 177 against Clade A and C virus respectively. In all cases, the paired FabA has significantly increased neutralization compared to FabG for all concentrations evaluated (Student’s t -test, p values ranging from p<0.01 to p<0.00001).
In summary, for all three HIV-1 clades, the CHla provides FabA with the capacity to neutralize CD4+ T-cell infection whereas the corresponding FabG harboring the same paratope but a CHly has limited neutralizing activities.
- Blockade of cell-to-cell virus transfer by FabA and FabG
Cell-to-cell transmission of HIV-1 is of major importance in vivo , being much more efficient than infection by cell-free virions [27] At the mucosal level, HIV-1 entry through multilayer mucosal tissues occurs mainly by targeting Langerhans cells (LCs) that in turn transfer to CD4+ T-cells [28] [29] [30] [31] that constitute the founder infected population. Moreover, CD4+T cells can disseminate out of the mucosal tissue and spread the virus to other CD4+T cells. Thus, we evaluated the capacity of the two pairs of ESN FabA and G to block clades A, B and C virus transfer from LCs to CD4+T-cells. Transfer of clade A and C HIV-1 is inhibited by both FabA 43 and 177 in a concentration dependent manner, as well as that of clade B by FabA 43, inducing a >40% inhibition at lOOng/ml. Transfer of clade B virus remains much less sensitive to FabA 177 . FabG 43 blocks transfer of Clade A and C viruses but less efficiently than its FabA counterpart and FabG 177 was only able to block clade A HIV-1 transfer. Of note, taken all the value together (including all viral clades and Fab isotypes) at lOOng/ml of Fab, neutralization activities correlated directly with inhibition of virus transfer (spearman r= 0,6655, p= 0,0219).
Taken together, this set of functional analyses demonstrate that switching the CHla from a mucosal Ab to CHly considerably reduces the Fab antiviral properties, either neutralization or HIV-1 transfer from LCs to CD4+T-cells.
1.6. FabA and G epitope mapping
As a further step in elucidating the molecular bases by which each specific CHI region impact on antibody affinity and functions, due to selective molecular stringency/flexibility imposed by CHI domain on the paratope conformation, we thought to determine the conformational epitopes specific for each Fab isotype. The molecular bases by which each specific CHI region impact on antibody affinity and functions could be due to selective molecular stringency/flexibility imposed by CHI domain on the paratope conformation. Thus, we thought to determine the conformational epitopes specific for each Fab isotype. Clones 43 and 177 specific FabA and FabG epitopes were characterized by combining epitope mapping, by screening a random peptide library, with an in-silico analysis. For this later, we used available clade B gp41 crystal structures, either in pre- or 6-Helix bundle conformations, that were further adapted by in silico modeling to model clade A and C gp41, as described in the method section.
Therefore, a 12mer random peptide library was used to characterize a set of the best specific peptides, referred to as mimotopes, for both 43 and 177 clones as FabA and FabG using three rounds of successive screening with increasing stringency, as we described [2]
After sequencing 100 phages specific for each of the Fabs, a larger set of peptides with higher occurrence were retrieved on each of the FabA compared to FabG, in line with the lower affinity of the FabGs compared with Fab As for gp41. We therefore concentrated our analysis below on the FabA 43 and 177 mimotopes. None of the selected mimotopes correspond to linear sequences of gp41, indicating that Fab epitopes are conformational, as already suggested [2]
To characterize these conformational epitopes, each set of FabA mimotopes was docked onto trimeric gp41 crystal structures using in silico approaches, as described in the method section and supplementary data. Only two gp41 crystal structures of a Clade B gp41 in different conformational states are available. One structure represents gp41 in the pre-fusion state together with gpl20, although this gp41 lacks the MPER region [32] The other gp41 structure mimics the post fusion state and is composed of three N-helices and three C-helices that form a six helix-bundle for completion of HIV- 1 fusion with target cells in a spring load model of fusion. In this case, the gp41 construct lacks the gp41 Cys- loop bridging the C and N helices [33]
Clone 43 is primarily specific for the gp41 MPER [2] and the Cys-loop contains important gp41 epitopes [34] Therefore, we first optimized the available crystal structure of gp41 clade B by reconstituting the missing MPER in the pre-fusion structure and the missing loop in the 6-Helix bundle as described in the supplementary data. As no crystal structure of clade A or C are accessible in the literature, we constructed A and clade C gp41 structure by mapping each of clade A and Clade C gp41 sequences onto the clade B gp41 structures, as detailed in the supplementary data. As a result, from these in silico modeling calculations, Clade A, B and C gp41 structures in the pre- and 6-Helix bundle conformation were available for epitope docking studies.
Selected FabA specific mimotopes obtained from FabA 43 and 177 were localized on clade A, B and C gp41 pre-fusion and six-helix bundle structures using the Pepsurf method as detailed in supplementary data. Three main regions were targeted by FabA 43 mimotopes on gp41 helix bundle structure of all three clades the highest hits being localized on the lower MPER region interface with the N-helix, in agreement with the screening strategy focusing on PI used for selecting FabA 43 [2] In comparison, FabA 43 mimotopes also fit on the pre-fusion structures, although in a more scattered manner and with lower scores. The main regions targeted by FabA 177 mimotopes on gp41 helix bundle structure of all three clades differ, as expected from the FabA 177 screening strategy, from those targeted by FabA 43 mimotopes, and the best fit appears in the loop linking the N and C gp41-helices. These FabA 177 mimotopes also match gp41 pre-fusion structures, but with lower scores and in a more scattered pattern, irrespective of the clade.
From these analysis, for each gp41 conformation and clade, an additional bioinformatic analysis described in the method section, allowed to characterized for FabA 43, 5 conformational epitopes that best mimic each set of mimotopes that could be used as a vaccine. Interestingly, only one of this conformational peptide was extracted from gp41 in a pre-fusion conformation, in agreement with lower scores obtained for mimotopes docking on pre-fusion versus 6-Helix bundle gp41 structure as shown above.
One out of 5 conformational epitopes defined in silico for FabA 43, referred to as P7, competed significantly with FabA 43 binding to PI and gp41/gpl40 of the three clades in ELISA when preincubated with the FabA 43 (Figure 1A) in a concentration-dependent manner (Figure IB). P7 at 5 mM could block FabA 43 binding by >50%, reaching >80% at 1 OmM (Figure IB). A 9 aa peptide derived from the influenza virus hemagglutinin (HA) used as negative control did not interfere with 177 binding in ELISA (Figure 1 A).
In contrast, peptide P7 did not interfere with Fab G 43 binding to the same antigens (not shown). Altogether, these results indicate that P7 interfere with FabA 43 cross-clade. P7 corresponds to an 11 aminoacid peptide located at the interface formed by the tips of Isl and H-helices of the three clades of gp41 (Figure 1C). This cross-clade activity is unexpected as P7 has been defined as the best conformational epitope extracted from in silico on FabA 43 mimotope fitting to the 6-Helix bundle Clade A gp41 structure. The other four conformational peptides defined in silico for FabA 43 that do not interfere with Fab A 43 binding to the gp41 antigens are located on different regions of gp41 on each gp41 clade, although for two of them spatially close from the region defined by.
Furthermore, at the functional level, preincubation of Fab A 43 with a 400- fold molar excess of P7 blocked the FabA 43 HIV-1 clade A, B and C neutralizing activities with an efficacy of 33+5, 54+8, 24+11 %, respectively (Figure IE). These results are in good agreement with the antigen binding blockade observed in ELISA (Figure 1 A, B).
The potential of P7 to inhibit CD4+T cells infection was then assessed in a single cycle infectivity assay. P7 induce inhibition of CD4+T cells infection in a dose-dependent manner (Figure 2). Finally we evaluate whether P7 could bind to HIV-1 envelope trimers gpl40 of clades A, B and C. Therefore, P7 at various concentrations (5, 10 and 20mM) or irrelevant HA peptide (20mM) were incubated with each immobilized gpl40. P7 binding was quantified using FabA 43 for detection (Figure 3A-B).
Results indicate that FabA 43 binds to each gpl40 clade in the absence (0 mM) and in the presence of 5 mM of P7. However, in the presence of 20 mM of P7, but not of HA peptide (not shown), Fab43 binding to each gpl40 clade increases by a factor 2. P7 being the specific epitope of FabA 43 and thus binding to it, as we already demonstrated (Khamassi et al, Plos Parth 2020), these results suggest a mechanism by which P7 binds to gpl40 increasing the epitope number accessible to the FabA. Furthermore, these results show that P7 binds the viral envelope cross clade, as does FabA 43 (Figure 3A-B).
REFERENCES
1. Miyazawa, M., et al., The 'immunologic advantage' of HIV-exposed seronegative individuals. Aids, 2009. 23(2): p. 161-75.
2. Tudor, D., et al., HIV-1 gp41-specific monoclonal mucosal IgAs derived from highly exposed but IgG- seronegative individuals block HIV-1 epithelial transcytosis and neutralize CD4(+) cell infection: an IgA gene and functional analysis. Mucosal Immunol, 2009. 2(5): p. 412-26.
3. Shen, R. and P.D. Smith, Mucosal correlates of protection in HIV-1- exposed sero-negative persons. Am J Reprod Immunol, 2014. 72(2): p. 219-27.
4. Lopez, E., et al., The multi-faceted nature of Immunoglobulin A and its complex role in HIV. AIDS Res Hum Retroviruses, 2018.
5. Yu, X., et al., Impact of IgA constant domain on HIV-1 neutralizing function of monoclonal antibody F425Alg8. J Immunol, 2013. 190(1): p. 205-10.
6. Clerici, M., et al, Serum IgA of HIV-exposed uninfected individuals inhibit HIV through recognition of a region within the alpha-helix of gp41. AIDS, 2002. 16(13): p. 1731-41.
7. Devito, C., et al, Cross-clade HIV- 1 -specific neutralizing IgA in mucosal and systemic compartments of HIV- 1 -exposed, persistently seronegative subjects. J Acquir Immune Defic Syndr, 2002. 30(4): p. 413-20.
8. Mouquet, H., Antibody B cell responses in HIV-1 infection. Trends Immunol, 2014. 35(11): p. 549-61.
9. Zwick, M.B., et al., The long third complementarity-determining region of the heavy chain is important in the activity of the broadly neutralizing anti-human immunodeficiency virus type 1 antibody 2F5. J Virol, 2004. 78(6): p. 3155-61.
10. Klein, F., et al, Somatic mutations of the immunoglobulin framework are generally required for broad and potent HIV-1 neutralization. Cell, 2013. 153(1): p. 126- 38. 11. Tudor, D., et al, Isotype modulates epitope specificity, affinity, and antiviral activities of anti-HIV- 1 human broadly neutralizing 2F5 antibody. Proc Natl Acad Sci U S A, 2012. 109(31): p. 12680-5.
12. Janda, A., et al., Ig Constant Region Effects on Variable Region Structure and Function. Front Microbiol, 2016. 7: p. 22.
13. Muster, T., et al., A conserved neutralizing epitope on gp41 of human immunodeficiency virus type 1. J Virol, 1993. 67(11): p. 6642-7.
14. Correa, A., et al., Structure of a human IgAl Fab fragment at 1.55 A resolution: potential effect of the constant domains on antigen-affinity modulation. Acta Crystallogr D Biol Crystallogr, 2013. 69(Pt 3): p. 388-97.
15. Crespillo, S., et al., Single-chain protein mimetics of the N-terminal heptad- repeat region of gp41 with potential as anti-HIV- 1 drugs. Proc Natl Acad Sci U S A, 2014. 111(51): p. 18207-12.
16. Sela-Culang, F, V. Kunik, and Y. Ofran, The structural basis of antibody- antigen recognition. Front Immunol, 2013. 4: p. 302.
17. Brandtzaeg, P., et al, Regional specialisation in the mucosal immune system: what happens in the microcomprtments? Immnunol. Today, 1999. 20(3): p. 141- 51.
18. Alfsen, A. and M. Bomsel, HIV-1 gp41 envelope residues 650-685 exposed on native virus act as a lectin to bind epithelial cell galactosyl ceramide. J Biol Chem, 2002. 277(28): p. 25649-59.
19. Bomsel, M., et al, Immunization with HIV-1 gp41 subunit virosomes induces mucosal antibodies protecting nonhuman primates against vaginal SHIV challenges. Immunity, 2011. 34(2): p. 269-80.
20. Kunert, R., et al, Characterization of molecular features, antigen-binding, and in vitro properties of IgG and IgM variants of 4E10, an anti-HIV type 1 neutralizing monoclonal antibody. AIDS Res Hum Retroviruses, 2004. 20(7): p. 755-62.
21. Bomsel, M., Transcytosis of infectious human immunodeficiency virus across a tight human epithelial cell line barrier. Nat Med, 1997. 3(1): p. 42-7. 22. Alfsen, A., et al., Secretory IgA specific for a conserved epitope on gp41 envelope glycoprotein inhibits epithelial transcytosis of HIV- 1. J Immunol, 2001. 166(10): p. 6257-65.
23. Magerus-Chatinet, A. and M. Bomsel, Galactosyl Ceramide is a common mucosal dendritic and epithelial cell receptor for HIV- 1 and mediates raft-dependent entry of HIV-1 across mucosa. Immunity, under review, 2004: p. 57-68.
24. Leroux-Roels, G., et al, Randomized Phase I: Safety, Immunogenicity and Mucosal Antiviral Activity in Young Healthy Women Vaccinated with HIV-1 Gp41 PI Peptide on Virosomes. PLoS One, 2013. 8(2): p. e55438.
25. Muster, T., et al, Cross-neutralizing activity against divergent human immunodeficiency virus type 1 isolates induced by the gp41 sequence ELDKWAS. J Virol, 1994. 68(6): p. 4031-4.
26. Mouquet, H., et al., Polyreactivity increases the apparent affinity of anti- HIV antibodies by heteroligation. Nature, 2010. 467(7315): p. 591-5.
27. Anderson, D.J., et al, Targeting Trojan Horse leukocytes for HIV prevention. AIDS, 2010. 24(2): p. 163-87.
28. Ganor, Y., et al., Within 1 h, HIV-1 uses viral synapses to enter efficiently the inner, but not outer, foreskin mucosa and engages Langerhans-T cell conjugates. Mucosal Immunol, 2010. 3(5): p. 506-22.
29. Hladik, F. and M.J. McElrath, Setting the stage: host invasion by HIV. Nat Rev Immunol, 2008. 8(6): p. 447-57.
30. Zhou, Z., et al, HIV-1 efficient entry in inner foreskin is mediated by elevated CCL5/RANTES that recruits T cells and fuels conjugate formation with Langerhans cells. PLoS Pathog, 2011. 7(6): p. el002100.
31. van den Berg, L.M. and T.B. Geijtenbeek, Antiviral immune responses by human langerhans cells and dendritic cells in HIV-1 infection. Adv Exp Med Biol, 2013. 762: p. 45-70.
32. Pancera, M., et al, Structure and immune recognition of trimeric pre-fusion HIV-1 Env. Nature, 2014. 514(7523): p. 455-61. 33. Buzon, V., et al, Crystal structure of HIV- 1 gp41 including both fusion peptide and membrane proximal external regions. PLoS Pathog, 2010. 6(5): p. el000880.
34. Ringel, O., et al, The Hard Way towards an Antibody-Based HIV-1 Env Vaccine: Lessons from Other Viruses. Viruses, 2018. 10(4).
35. Briney, B.S., et al., Tissue-specific expressed antibody variable gene repertoires. PLoS One, 2014. 9(6): p. el00839.
36. Bruzzoni-Giovanelli, H., et al, Interfering peptides targeting protein-protein interactions: the next generation of drugs? Drug Discov Today, 2018. 23(2): p. 272-285.
37. Sanders, R.W. and J.P. Moore, HIV: A stamp on the envelope. Nature, 2014. 514(7523): p. 437-8.
38. Mkhize, N.N., et al., Broadly neutralizing antibody specificities detected in the genital tract of HIV-1 infected women. AIDS, 2016. 30(7): p. 1005-14.
39. Buonaguro, L., M.L. Tomesello, and F.M. Buonaguro, Human immunodeficiency virus type 1 subtype distribution in the worldwide epidemic: pathogenetic and therapeutic implications. J Virol, 2007. 81(19): p. 10209-19.
40. Lai, R.P., et al, A fusion intermediate gp41 immunogen elicits neutralizing antibodies to HIV-1. J Biol Chem, 2014. 289(43): p. 29912-26.
41. Fiedler, M. and A. Skerra, Use of thiophilic adsorption chromatography for the one-step purification of a bacterially produced antibody F(ab) fragment without the need for an affinity tag. Protein Expr Purif, 1999. 17(3): p. 421-7.
42. Frey, G., et al., A fusion-intermediate state of HIV-1 gp41 targeted by broadly neutralizing antibodies. Proc Natl Acad Sci U S A, 2008. 105(10): p. 3739-44.
43. Duchemin, M., et al., IgA Targeting Human Immunodeficiency Virus-1 Envelope gp41 Triggers Antibody-Dependent Cellular Cytotoxicity Cross-Clade and Cooperates with gp41-Specific IgGto Increase Cell Lysis. Front Immunol, 2018. 9: p. 244.
44. Weissenhorn, W., et al, Assembly of a rod-shaped chimera of a trimeric GCN4 zipper and the HIV-1 gp41 ectodomain expressed in Escherichia coli. Proc Natl Acad Sci U S A, 1997. 94(12): p. 6065-9. 45. Caffrey, M., et al, Three-dimensional solution structure of the 44 kDa ectodomain of SIV gp41. EMBO J, 1998. 17(16): p. 4572-84.
46. Webb, B. and A. Sali, Protein structure modeling with MODELLER. Methods Mol Biol, 2014. 1137: p. 1-15.
47. Sen, J., et al, The disulfide loop of gp41 is critical to the furin recognition site of HIV gpl60. Protein Sci, 2007. 16(6): p. 1236-41.
48. Caffrey, M., Model for the structure of the HIV gp41 ectodomain: insight into the intermolecular interactions of the gp41 loop. Biochim Biophys Acta, 2001. 1536(2-3): p. 116-22.
49. Guyon, F., et al., BCSearch: fast structural fragment mining over large collections of protein structures. Nucleic Acids Res, 2015. 43(W1): p. W378-82.
50. Hess, B., et al., GROMACS 4: Algorithms for Highly Efficient, Load- Balanced, and Scalable Molecular Simulation. J Chem Theory Comput, 2008. 4(3): p. 435- 47.
51. Pronk, S., et al, GROMACS 4.5: a high-throughput and highly parallel open source molecular simulation toolkit. Bioinformatics, 2013. 29(7): p. 845-54.
52. Lindorff-Larsen, K., et al., Improved side-chain torsion potentials for the Amber ff99SB protein force field. Proteins, 2010. 78(8): p. 1950-8.
53. Jorgensen, W.L., J. Chandrasekhar, and J.D. Madura, Comparison of simple potential functions for simulating liquid water J. Chem. Phys., 1983. 79(2): p. 926-936.
54. Rubinstein, N.D., et al, Computational characterization of B-cell epitopes. Mol Immunol, 2008. 45(12): p. 3477-89.
55. Pacios, L.F., et al., LocaPep: localization of epitopes on protein surfaces using peptides from phage display libraries. J Chem Inf Model, 2011. 51(6): p. 1465-73.
56. Lamiable, A., et al., PEP-FOLD3: faster de novo structure prediction for linear peptides in solution and in complex. Nucleic Acids Res, 2016. 44(W1): p. W449-54.

Claims

1. An isolated peptide comprising or consisting of the consecutive sequence of amino acids of sequence: LWNWFDISAASI (SEQ IDN°1).
2. A therapeutic composition comprising a peptide according to claim 1 for the preventive or curative treatment of an HIV infection.
3. A vaccine composition comprising a peptide according to claim 1, or a nucleic acid encoding said peptide.
4. A composition according to claim 2 or 3, which is suitable for mucosal administration.
5. A vaccine composition according to claim 3, further comprising an adjuvant.
6. A peptide of claim 1 or a composition of any one of claims 2 to 5 for use in the preventive treatment of HIV infection or AIDS.
7. A peptide of claim 1 or a composition of any one of claims 2 to 4 for use in a method of inducing a mucosal immune response to HIV-1.
8. A peptide according to claim 1, or a nucleic acid encoding said peptide, for use in a method of inducing a mucosal IgA response to HIV-1, wherein said peptide has the sequence SEQ ID N° 1.
9. A peptide according to claim 1, or a nucleic acid encoding said peptide, for use in a method of inducing a mucosal IgG response to HIV-1, wherein said peptide has the sequence SEQ ID N° 1.
10 An isolated nucleic acid encoding the amino acid sequence of a peptide according to claim 1.
11. A method of prevention or treatment of an HIV infection, comprising administration of a composition of any one of claims 2 to 5 to an individual in need thereof.
PCT/EP2021/056710 2020-03-17 2021-03-16 Vaccine compositions for hiv prevention and treatment WO2021185851A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP20305278 2020-03-17
EP20305278.2 2020-03-17

Publications (1)

Publication Number Publication Date
WO2021185851A1 true WO2021185851A1 (en) 2021-09-23

Family

ID=71111377

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2021/056710 WO2021185851A1 (en) 2020-03-17 2021-03-16 Vaccine compositions for hiv prevention and treatment

Country Status (1)

Country Link
WO (1) WO2021185851A1 (en)

Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4744984A (en) 1985-10-08 1988-05-17 Vetrepharm Research, Inc. Antiviral immunotherapeutic agent and preparation thereof
WO2001047955A2 (en) 1999-12-23 2001-07-05 Medical Research Council Improvements in or relating to immune responses to hiv
US6455265B1 (en) 1997-11-17 2002-09-24 Mymetics S.A. Method for obtaining vaccines for preventing the pathogenic effects related to a retroviral infection
WO2004085466A2 (en) * 2003-03-28 2004-10-07 The Government Of The United States Of America, As Represented By The Secretary Of The Department Of Health And Human Services, Centers For Disease Control And Prevention Immunogenic hiv-1 multi-clade, multivalent constructs and methods of their use
WO2005010033A1 (en) 2003-07-30 2005-02-03 Mymetics Corporation New soluble and stabilized trimeric form of gp41 polypeptides
WO2007107597A2 (en) * 2006-03-21 2007-09-27 Bundesrepublik Deutschland, Vertreten Durch Das Bundesministerium Für Gesundheit, Dieses Vertreten Durch Das Robert-Koch-Institut Immunogenic construct and a method for the prophylactic or therapeutic treatment of aids
WO2009021230A1 (en) * 2007-08-09 2009-02-12 University Of Medicine And Dentistry Of New Jersey Hiv-1 gp41 neutralization domain and use thereof
US20110124842A1 (en) * 2003-09-19 2011-05-26 Brunel Florence M Peptide that binds to a broadly neutralizing anti-HIV antibody-structure of 4E10 Fab fragment complex, uses thereof, compositions therefrom

Patent Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4744984A (en) 1985-10-08 1988-05-17 Vetrepharm Research, Inc. Antiviral immunotherapeutic agent and preparation thereof
US6455265B1 (en) 1997-11-17 2002-09-24 Mymetics S.A. Method for obtaining vaccines for preventing the pathogenic effects related to a retroviral infection
WO2001047955A2 (en) 1999-12-23 2001-07-05 Medical Research Council Improvements in or relating to immune responses to hiv
WO2004085466A2 (en) * 2003-03-28 2004-10-07 The Government Of The United States Of America, As Represented By The Secretary Of The Department Of Health And Human Services, Centers For Disease Control And Prevention Immunogenic hiv-1 multi-clade, multivalent constructs and methods of their use
WO2005010033A1 (en) 2003-07-30 2005-02-03 Mymetics Corporation New soluble and stabilized trimeric form of gp41 polypeptides
US20110124842A1 (en) * 2003-09-19 2011-05-26 Brunel Florence M Peptide that binds to a broadly neutralizing anti-HIV antibody-structure of 4E10 Fab fragment complex, uses thereof, compositions therefrom
WO2007107597A2 (en) * 2006-03-21 2007-09-27 Bundesrepublik Deutschland, Vertreten Durch Das Bundesministerium Für Gesundheit, Dieses Vertreten Durch Das Robert-Koch-Institut Immunogenic construct and a method for the prophylactic or therapeutic treatment of aids
WO2009021230A1 (en) * 2007-08-09 2009-02-12 University Of Medicine And Dentistry Of New Jersey Hiv-1 gp41 neutralization domain and use thereof

Non-Patent Citations (64)

* Cited by examiner, † Cited by third party
Title
"Remington: The Science and Practice of Pharmacy", 2005, LIPPINCOT AND WILLIAMS
ALFSEN, A. ET AL.: "Secretory IgA specific for a conserved epitope on gp41 envelope glycoprotein inhibits epithelial transcytosis of HIV-1", J IMMUNOL, vol. 166, no. 10, 2001, pages 6257 - 65, XP002336811
ALFSEN, A.M. BOMSEL: "HIV-1 gp41 envelope residues 650-685 exposed on native virus act as a lectin to bind epithelial cell galactosyl ceramide", J BIOL CHEM, vol. 277, no. 28, 2002, pages 25649 - 59
ANDERSON, D.J. ET AL.: "Targeting Trojan Horse leukocytes for HIV prevention", AIDS, vol. 24, no. 2, 2010, pages 163 - 87
BITTER ET AL., CURR PROBL DERMATOL, vol. 40, 2011, pages 20 - 35
BOMSEL, M. ET AL.: "Immunization with HIV-1 gp41 subunit virosomes induces mucosal antibodies protecting nonhuman primates against vaginal SHIV challenges", IMMUNITY, vol. 34, no. 2, 2011, pages 269 - 280, XP028176138, DOI: 10.1016/j.immuni.2011.01.015
BOMSEL, M.: "Transcytosis of infectious human immunodeficiency virus across a tight human epithelial cell line barrier", NAT MED, vol. 3, no. 1, 1997, pages 42 - 7
BRANDTZAEG, P. ET AL.: "Regional specialisation in the mucosal immune system: what happens in the microcomprtments?", IMMNUNOL. TODAY, vol. 20, no. 3, 1999, pages 141 - 51
BRANDZTAEG, AJRCCM, 2011
BRINEY, B.S. ET AL.: "Tissue-specific expressed antibody variable gene repertoires", PLOS ONE, vol. 9, no. 6, 2014, pages el00839
BRUZZONI-GIOVANELLI, H. ET AL.: "Interfering peptides targeting protein-protein interactions: the next generation of drugs?", DRUG DISCOV TODAY, vol. 23, no. 2, 2018, pages 272 - 285
BUONAGURO, L.M.L. TORNESELLOF.M. BUONAGURO: "Human immunodeficiency virus type 1 subtype distribution in the worldwide epidemic: pathogenetic and therapeutic implications", J VIROL, vol. 81, no. 19, 2007, pages 10209 - 19, XP055331623, DOI: 10.1128/JVI.00872-07
BUZON, V. ET AL.: "Crystal structure of HIV-1 gp41 including both fusion peptide and membrane proximal external regions", PLOS PATHOG, vol. 6, no. 5, 2010, pages el000880
CAFFREY, M. ET AL.: "Three-dimensional solution structure of the 44 kDa ectodomain of SIV gp41", EMBO J, vol. 17, no. 16, 1998, pages 4572 - 84, XP002235688, DOI: 10.1093/emboj/17.16.4572
CAFFREY, M.: "Model for the structure of the HIV gp41 ectodomain: insight into the intermolecular interactions of the gp41 loop", BIOCHIM BIOPHYS ACTA, vol. 1536, no. 2-3, 2001, pages 116 - 22, XP004245564, DOI: 10.1016/S0925-4439(01)00042-4
CLERICI, M. ET AL.: "Serum IgA of HIV-exposed uninfected individuals inhibit HIV through recognition of a region within the alpha-helix of gp41", AIDS, vol. 16, no. 13, 2002, pages 1731 - 41, XP009022167, DOI: 10.1097/00002030-200209060-00004
CORREA, A. ET AL.: "Structure of a human IgA1 Fab fragment at 1.55 A resolution: potential effect of the constant domains on antigen-affinity modulation", ACTA CRYSTALLOGR D BIOL CRYSTALLOGR, vol. 69, 2013, pages 388 - 97
CRESPILLO, S. ET AL.: "Single-chain protein mimetics of the N-terminal heptad-repeat region of gp41 with potential as anti-HIV-1 drugs", PROC NATL ACAD SCI U S A, vol. 111, no. 51, 2014, pages 18207 - 12
D TUDOR ET AL: "HIV-1 gp41-specific monoclonal mucosal IgAs derived from highly exposed but IgG-seronegative individuals block HIV-1 epithelial transcytosis and neutralize CD4+ cell infection: an IgA gene and functional analysis", MUCOSAL IMMUNOLOGY, vol. 2, no. 5, 1 September 2009 (2009-09-01), US, pages 412 - 426, XP055717138, ISSN: 1933-0219, DOI: 10.1038/mi.2009.89 *
DEVITO, C. ET AL.: "Cross-clade HIV-1-specific neutralizing IgA in mucosal and systemic compartments of HIV-1-exposed, persistently seronegative subjects", J ACQUIR IMMUNE DEFIC SYNDR, vol. 30, no. 4, 2002, pages 413 - 20
DUCHEMIN, M. ET AL.: "IgA Targeting Human Immunodeficiency Virus-1 Envelope gp41 Triggers Antibody-Dependent Cellular Cytotoxicity Cross-Clade and Cooperates with gp41-Specific IgGto Increase Cell Lysis", FRONT IMMUNOL, vol. 9, 2018, pages 244
FIEDLER, M.A. SKERRA: "Use of thiophilic adsorption chromatography for the one-step purification of a bacterially produced antibody F(ab) fragment without the need for an affinity tag", PROTEIN EXPR PURIF, vol. 17, no. 3, 1999, pages 421 - 7, XP004441591, DOI: 10.1006/prep.1999.1142
FREY, G. ET AL.: "A fusion-intermediate state of HIV-1 gp41 targeted by broadly neutralizing antibodies", PROC NATL ACAD SCI USA, vol. 105, no. 10, 2008, pages 3739 - 44, XP055291363, DOI: 10.1073/pnas.0800255105
GANOR, Y. ET AL.: "Within 1 h, HIV-1 uses viral synapses to enter efficiently the inner, but not outer, foreskin mucosa and engages Langerhans-T cell conjugates", MUCOSAL IMMUNOL, vol. 3, no. 5, 2010, pages 506 - 22
GUYON, F. ET AL.: "BCSearch: fast structural fragment mining over large collections of protein structures", NUCLEIC ACIDS RES, vol. 43, no. W1, 2015, pages W378 - 82
HESS, B. ET AL.: "GROMACS 4: Algorithms for Highly Efficient, Load-Balanced, and Scalable Molecular Simulation", J CHEM THEORY COMPUT, vol. 4, no. 3, 2008, pages 435 - 47, XP055441952, DOI: 10.1021/ct700301q
HLADIK, F.M.J. MCELRATH: "Setting the stage: host invasion by HIV", NAT REV IMMUNOL, vol. 8, no. 6, 2008, pages 447 - 57, XP037115114, DOI: 10.1038/nri2302
J. D. NELSON ET AL: "An Affinity-Enhanced Neutralizing Antibody against the Membrane-Proximal External Region of Human Immunodeficiency Virus Type 1 gp41 Recognizes an Epitope between Those of 2F5 and 4E10", JOURNAL OF VIROLOGY, vol. 81, no. 8, 15 April 2007 (2007-04-15), US, pages 4033 - 4043, XP055328826, ISSN: 0022-538X, DOI: 10.1128/JVI.02588-06 *
JANDA, A. ET AL.: "Ig Constant Region Effects on Variable Region Structure and Function", FRONT MICROBIOL, vol. 7, 2016, pages 22
JORGENSEN, W.L.J. CHANDRASEKHARJ.D. MADURA: "Comparison of simple potential functions for simulating liquid water", J. CHEM. PHYS., vol. 79, no. 2, 1983, pages 926 - 936
KHAMASSI ET AL., PLOS PARTH, 2020
KLEIN, F. ET AL.: "Somatic mutations of the immunoglobulin framework are generally required for broad and potent HIV-1 neutralization", CELL, vol. 153, no. 1, 2013, pages 126 - 38, XP055119651, DOI: 10.1016/j.cell.2013.03.018
KUNERT, R. ET AL.: "Characterization of molecular features, antigen-binding, and in vitro properties of IgG and IgM variants of 4E10, an anti-HIV type 1 neutralizing monoclonal antibody", AIDS RES HUM RETROVIRUSES, vol. 20, no. 7, 2004, pages 755 - 62
LAI, R.P. ET AL.: "A fusion intermediate gp41 immunogen elicits neutralizing antibodies to HIV-1", J BIOL CHEM, vol. 289, no. 43, 2014, pages 29912 - 26, XP055507267, DOI: 10.1074/jbc.M114.569566
LAMIABLE, A. ET AL.: "PEP-FOLD3: faster de novo structure prediction for linear peptides in solution and in complex", NUCLEIC ACIDS RES, vol. 44, no. W1, 2016, pages W449 - 54
LEE ET AL., VACCINE, vol. 29, 2011, pages 417 - 425
LEROUX-ROELS, G. ET AL.: "Randomized Phase I: Safety, Immunogenicity and Mucosal Antiviral Activity in Young Healthy Women Vaccinated with HIV-1 Gp41 PI Peptide on Virosomes", PLOS ONE, vol. 8, no. 2, 2013, pages e55438
LINDORFF-LARSEN, K. ET AL.: "Improved side-chain torsion potentials for the Amber ff99SB protein force field", PROTEINS, vol. 78, no. 8, 2010, pages 1950 - 8
LOPEZ, E. ET AL.: "The multi-faceted nature of Immunoglobulin A and its complex role in HIV", AIDS RES HUM RETROVIRUSES, 2018
MAGERUS-CHATINET, A.M. BOMSEL: "Galactosyl Ceramide is a common mucosal dendritic and epithelial cell receptor for HIV-1 and mediates raft-dependent entry of HIV-1 across mucosa", IMMUNITY, 2004, pages 57 - 68
MIYAZAWA, M. ET AL.: "The 'immunologic advantage' of HIV-exposed seronegative individuals", AIDS, vol. 23, no. 2, 2009, pages 161 - 75
MKHIZE, N.N. ET AL.: "Broadly neutralizing antibody specificities detected in the genital tract of HIV-1 infected women", AIDS, vol. 30, no. 7, 2016, pages 1005 - 14
MOUQUET, H. ET AL.: "Polyreactivity increases the apparent affinity of anti-HIV antibodies by heteroligation", NATURE, vol. 467, no. 7315, 2010, pages 591 - 5, XP055242135, DOI: 10.1038/nature09385
MOUQUET, H.: "Antibody B cell responses in HIV-1 infection", TRENDS IMMUNOL, vol. 35, no. 11, 2014, pages 549 - 61
MUSTER, T. ET AL.: "A conserved neutralizing epitope on gp41 of human immunodeficiency virus type 1", J VIROL, vol. 67, no. 11, 1993, pages 6642 - 7, XP000613016
MUSTER, T. ET AL.: "Cross-neutralizing activity against divergent human immunodeficiency virus type 1 isolates induced by the gp41 sequence ELDKWAS", J VIROL, vol. 68, no. 6, 1994, pages 4031 - 4
PACIOS, L.F. ET AL.: "LocaPep: localization of epitopes on protein surfaces using peptides from phage display libraries", J CHEM INF MODEL, vol. 51, no. 6, 2011, pages 1465 - 73
PANCERA, M. ET AL.: "Structure and immune recognition of trimeric pre-fusion HIV-1 Env", NATURE, vol. 514, no. 7523, 2014, pages 455 - 61, XP055178485, DOI: 10.1038/nature13808
PRONK, S. ET AL.: "GROMACS 4.5: a high-throughput and highly parallel open source molecular simulation toolkit", BIOINFORMATICS, vol. 29, no. 7, 2013, pages 845 - 54
RINGEL, O. ET AL.: "The Hard Way towards an Antibody-Based HIV-1 Env Vaccine: Lessons from Other Viruses", VIRUSES, vol. 10, no. 4, 2018
RUBINSTEIN, N.D. ET AL.: "Computational characterization of B-cell epitopes", MOL IMMUNOL, vol. 45, no. 12, 2008, pages 3477 - 89, XP022709840, DOI: 10.1016/j.molimm.2007.10.016
SANDERS, R.W.J.P. MOORE: "HIV: A stamp on the envelope", NATURE, vol. 514, no. 7523, 2014, pages 437 - 8
SCHIJNS ET AL., CURR. OPIN. IMMUNOL, vol. 12, 2000, pages 456
SELA-CULANG, I.V. KUNIKY. OFRAN: "The structural basis of antibody-antigen recognition", FRONT IMMUNOL, vol. 4, 2013, pages 302, XP055557261, DOI: 10.3389/fimmu.2013.00302
SEN, J. ET AL.: "The disulfide loop of gp41 is critical to the furin recognition site of HIV gpl60", PROTEIN SCI, vol. 16, no. 6, 2007, pages 1236 - 41
SHEN, R.P.D. SMITH: "Mucosal correlates of protection in HIV-1-exposed sero-negative persons", AM J REPROD IMMUNOL, vol. 72, no. 2, 2014, pages 219 - 27
TUDOR, D. ET AL.: "HIV-1 gp41-specific monoclonal mucosal IgAs derived from highly exposed but IgG-seronegative individuals block HIV-1 epithelial transcytosis and neutralize CD4(+) cell infection: an IgA gene and functional analysis", MUCOSAL IMMUNOL, vol. 2, no. 5, 2009, pages 412 - 26, XP055717138, DOI: 10.1038/mi.2009.89
TUDOR, D. ET AL.: "Isotype modulates epitope specificity, affinity, and antiviral activities of anti-HIV-1 human broadly neutralizing 2F5 antibody", PROC NATL ACAD SCI USA, vol. 109, no. 31, 2012, pages 12680 - 5, XP055055874, DOI: 10.1073/pnas.1200024109
VAN DEN BERG, L.M.T.B. GEIJTENBEEK: "Antiviral immune responses by human langerhans cells and dendritic cells in HIV-1 infection", ADV EXP MED BIOL, vol. 762, 2013, pages 45 - 70
WEBB, B.A. SALI: "Protein structure modeling with MODELLER", METHODS MOL BIOL, vol. 1137, 2014, pages 1 - 15
WEISSENHORN, W. ET AL.: "Assembly of a rod-shaped chimera of a trimeric GCN4 zipper and the HIV-1 gp41 ectodomain expressed in Escherichia coli", PROC NATL ACAD SCI USA, vol. 94, no. 12, 1997, pages 6065 - 9, XP002939089, DOI: 10.1073/pnas.94.12.6065
YU, X. ET AL.: "Impact of IgA constant domain on HIV-1 neutralizing function of monoclonal antibody F425Alg8", J IMMUNOL, vol. 190, no. 1, 2013, pages 205 - 10, XP055245717, DOI: 10.4049/jimmunol.1201469
ZHOU, Z. ET AL.: "HIV-1 efficient entry in inner foreskin is mediated by elevated CCL5/RANTES that recruits T cells and fuels conjugate formation with Langerhans cells", PLOS PATHOG, vol. 7, no. 6, 2011, pages el002100
ZWICK, M.B. ET AL.: "The long third complementarity-determining region of the heavy chain is important in the activity of the broadly neutralizing anti-human immunodeficiency virus type 1 antibody 2F5", J VIROL, vol. 78, no. 6, 2004, pages 3155 - 61, XP008125658, DOI: 10.1128/JVI.78.6.3155-3161.2004

Similar Documents

Publication Publication Date Title
JP7126594B2 (en) human immunodeficiency virus neutralizing antibody
US11053285B2 (en) Nucleic acids encoding human immunodeficiency virus type 1 (HIV-1) N-terminal deleted gp120 immunogens and methods of use
ES2789348T3 (en) Neutralizing antibodies to GP120 and their uses
WO2016196975A1 (en) Neutralizing antibodies to hiv-1 env and their use
US20150175678A1 (en) IgA CD4i ANTIBODIES AND METHODS OF TREATMENT USING SAME
WO2013085550A2 (en) V1v2 immunogens
JP2013520498A (en) Methods for inducing production of protective anti-HIV-1 antibodies
US20210292396A1 (en) Enginerred antibodies to hiv env
US20120121633A1 (en) Hiv cd4 binding site based covalent immunogen compositions
US9775895B2 (en) HIV therapeutics and methods of making and using same
CA2832148A1 (en) Peptide which can induce antibody capable of recognizing stereostructure of hiv
WO2021185851A1 (en) Vaccine compositions for hiv prevention and treatment
US20150004190A1 (en) Vaccine compositions for hiv prevention and treatment
JP2010509340A (en) Binary epitope antibody and B cell superantigen immunostimulator
Tan et al. The membrane-proximal region of C–C chemokine receptor type 5 participates in the infection of HIV-1
CA2562385A1 (en) Identification of the precise amino acid sequence of the epitope recognized by the potent neutralizing human anti-hiv-1 monoclonal antibody igg1b12
US9944693B2 (en) HIV antigens and antibodies and compositions, uses and methods thereof
US20230134571A1 (en) Engineered antibodies to hiv env
Omorodion Broadly Neutralizing Antibodies to HIV: Evolution of Structure in Three Infected Donors
AU2015215643B2 (en) A new non-HIV vaccine antigen from the vaginal microbiota capable of inducing a mucosal neutralizing protective antibody response against HIV infection
WO2023192881A1 (en) Neutralizing antibodies to hiv-1 env and their use
US20080146499A1 (en) Identification of the Precise Amino Acid Sequence of the Epitope Recognized by the Potent Neutralizing Human Anti-Hiv-1 Monoclonal Antibody Igg1b12
Morales V1/V2 domain scaffolds to improve the magnitude and quality of protective antibody responses to HIV-1
Liu Antigen-Binding Fragments: Production for and Use in Crystallographic Studies

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 21711289

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 21711289

Country of ref document: EP

Kind code of ref document: A1